Modeling, Synthesis, and Biological Evaluation of Potential Retinoid X

Nov 1, 2013 - ... (RAR) compared to all-trans-retinoic acid, with select analogues also reducing inhibition of other RXR-dependent pathways (e.g., VDR...
0 downloads 0 Views 1MB Size
Article pubs.acs.org/jmc

Modeling, Synthesis, and Biological Evaluation of Potential Retinoid X Receptor (RXR) Selective Agonists: Novel Analogues of 4‑[1(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydro-2naphthyl)ethynyl]benzoic Acid (Bexarotene) and (E)‑3-(3-(1,2,3,4tetrahydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)-4hydroxyphenyl)acrylic Acid (CD3254) Peter W. Jurutka,†,∥ Ichiro Kaneko,†,∥ Joanna Yang,† Jaskaran S. Bhogal,† Johnathon C. Swierski,† Christa R. Tabacaru,† Luis A. Montano,† Chanh C. Huynh,† Rabia A. Jama,† Ryan D. Mahelona,† Joseph T. Sarnowski,† Lisa M. Marcus,† Alexis Quezada,† Brittney Lemming,† Maria A. Tedesco,† Audra J. Fischer,† Said A. Mohamed,† Joseph W. Ziller,‡ Ning Ma,§ Geoffrey M. Gray,§ Arjan van der Vaart,§ Pamela A. Marshall,† and Carl E. Wagner*,† †

School of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, 4701 West Thunderbird Road, Glendale, Arizona 85306, United States ‡ Department of Chemistry, University of California, Irvine, 576 Rowland Hall, Irvine, California 92697, United States § Department of Chemistry, University of South Florida, 4202 East Fowler Avenue, CHE 205, Tampa, Florida, 33620, United States S Supporting Information *

ABSTRACT: Three unreported analogues of 4-[1-(3,5,5,8,8pentamethyl-5-6-7-8-tetrahydro-2-naphthyl)ethynyl]benzoic acid (1), otherwise known as bexarotene, as well as four novel analogues of (E)-3-(3-(1,2,3,4-tetrahydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)-4-hydroxyphenyl)acrylic acid (CD3254), are described and evaluated for their retinoid X receptor (RXR) selective agonism. Compound 1 has FDA approval as a treatment for cutaneous T-cell lymphoma (CTCL), although treatment with 1 can elicit side-effects by disrupting other RXR-heterodimer receptor pathways. Of the seven modeled novel compounds, all analogues stimulate RXR-regulated transcription in mammalian 2 hybrid and RXRE-mediated assays, possess comparable or elevated biological activity based on EC50 profiles, and retain similar or improved apoptotic activity in CTCL assays compared to 1. All novel compounds demonstrate selectivity for RXR and minimal crossover onto the retinoic acid receptor (RAR) compared to all-trans-retinoic acid, with select analogues also reducing inhibition of other RXR-dependent pathways (e.g., VDR-RXR). Our results demonstrate that further improvements in biological potency and selectivity of bexarotene can be achieved through rational drug design.



INTRODUCTION The human retinoid X receptors (hRXRs) consist of three identified isoforms (α, β, γ)1 that function as transcription factors, often in partnership with other members of a larger nuclear receptor (NR) superfamily of transcriptional regulators including the thyroid hormone receptor (TR), the vitamin D receptor (VDR), the liver X receptor (LXR), the peroxisome proliferator-activated receptor (PPAR), and the retinoic acid receptor (RAR), to name a few. All of these nuclear receptors generally promote or modulate gene transcription in the presence of the appropriate receptor ligand. This ligand, usually comprising an endogenous molecule, binds to the receptor ligand-binding domain (LBD), effecting a conformational shift that enables the receptor to associate with high affinity to its © 2013 American Chemical Society

specific hormone responsive element (HRE) on DNA. While an increasing number of HREs have been located at a considerable distance either upstream or downstream from their regulated genes, many HREs are often found within or close to the promoter region of the regulated gene. Typically, HREs consist of minimal core hexad sequences comprising two half-sites punctuated by a nucleotide spacer of variable length between direct, inverted, or everted repeats.2 Nuclear receptor proteins, as homodimers or heterodimers, regulate transcription when binding to their corresponding half-sites within the HREs. Received: June 10, 2013 Published: November 1, 2013 8432

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

benzoic acid (1)17 was shown to be a highly specific RXR agonist that Ligand Pharmaceuticals Inc. further developed to become an FDA approved drug, commonly known as bexarotene, for treatment of cutaneous T-cell lymphoma. Several analogues of bexarotene, some quite similar such as the disilabexarotene (2),18 have since been reported and shown to possess similarly specific RXR agonism.

While RAR, VDR, and TR were initially thought to bind to their HREs as homodimers,3 they actually bind as heterodimers with RXR.4 When RXR binds to its endogenous 9-cis-retinoic acid (9-cis-RA) agonist ligand, an RXR homodimer forms that subsequently binds to the RXR responsive element (RXRE). However, when RXR operates as a heterodimeric partner with other nuclear receptors, it can do so either in the presence or absence of a ligand in the RXR LBD. For the RXR-VDR heterodimer, evidence suggests that RXR is unoccupied.5 Alternatively, data suggest that RXR may possess a ligand when it operates as a heterodimer with LXR.6 Notably, RXR has been described as the central NR regulator because it often plays a critical role in heterodimer complex formation with the other NRs to allow DNA binding to their respective response elements.7 These and other observations employing a number of studies with RXR, rexinoids, and a wide range of nuclear receptors, have led to the classification of at least two types of RXRheterodimer complexes. These have been termed nonpermissive or permissive heterodimers. Purely nonpermissive heterodimers can only be activated by the ligand of the RXR partner receptor, while permissive heterodimers that can be activated either by an RXR ligand or by a partner-specific ligand.8 The nonpermissive heterodimers include RXR−TR, RXR−VDR, and RXR−RAR. Under most (but not all) conditions, RXR is completely “silent” in TR and VDR heterodimers, while RXR−RAR heterodimers can be further activated by select RXR ligands in the presence of an RAR ligand. Furthermore, in some cases, unique rexinoids have also been identified that stimulate RXR−RAR, even in the absence of a RAR ligand.9 Thus, RXR−RAR can also be considered conditionally nonpermissive. In contrast, the fully permissive heterodimers include the RXR−LXRs, RXR−PPARs, and RXR−FXR. The development of RXR-specific ligands for therapeutic use is complicated by the observation that not only are some RXR heterodimers permissive but also that under certain cellular conditions, or in particular cell types, the pool of RXR may be somewhat limiting. This has led to the observation that RXR ligands, like 9-cis RA for example, can inhibit nonpermissive nuclear receptor transactivation by such receptors as VDR10 and TR.11 Moreover, nuclear receptor ligands like thyroid hormone or 1,25-dihydroxyvitamin D3 (1,25D) that stimulate RXR heterodimerization with their cognate receptor are thought to participate in crosstalk inhibition of signaling by other receptors that also utilize the limited intracellular RXR pool, and this has been reported for T3−TR−RXR-mediated inhibition of VDR transactivation12 and 1,25D−VDR−RXRdirected inhibition of TR activity12b,13 although the mechanism involved in cross-receptor squelching likely extends beyond just sequestration of RXR. Nonetheless, a central issue in the development of novel therapeutic rexinoids is the creation of compounds that are likely to achieve a greater degree of heterodimer selectivity. Thus, changing the structure of the central RXR heteropartner ligand may result in specific NR modulators (SNuRMs) that possess unique characteristics that may exert a novel combination of NR control.14 RXR selective ligand (rexinoid) SNuRMs present attractive medicinal targets because selectively activating RXR versus RAR confers chemotherapeutic effects for several human cancers without incurring negative crossover RAR effects.15 Following lengthy synthesis and testing16 of molecules modeled after 9-cis RA, 4-[1-(3,5,5,8,8-pentamethyltetralin-2-yl)ethynyl]-

While 1 is primarily used to treat CTCL, it has also been explored as a potential treatment for colon cancer19 and breast cancer,20 and it has even been applied as an off-label treatment for lung cancer21 because promoting the transcription of RXRregulated genes seems to suppress uncontrolled cell proliferation and predispose cancer cells to undergo apoptosis during chemotherapy.20 Several RXR selective agonists, including compound 1, have been explored in mouse models of noninsulin-dependent diabetes mellitus (NIDDM) because of the ability of RXR to partner with PPAR.22 Despite the specificity that compound 1 displays for RXR in comparison to RAR, the known side effects of 1 include hypothyroidism,23 cutaneous toxicity, and hyperlipidemia. These side effects of 1 are likely incurred either by nonpermissive receptor antagonism, as has been noted for TR24 to explain RXR agonist induced hypothyroidism, or permissive receptor agonism, such as LXR to effect hyperlipidemia25 or RAR to effect cutaneous toxicity26 at typical dose concentrations. Thus, there is compelling motivation to develop novel RXR selective agonists whose effects on other receptor mediated pathways are attenuated or altogether absent. In addition to this motivation, several RXR agonists are being studied to treat other diseases through RXR-impacted pathways, now including two separate clinical trials of 1 in Alzheimer’s disease (AD) based on upregulation of apoE in mouse models of AD.27 There are several RXR specific agonists modeled on 9-cis RA and 1 reported in literature. For example, the cyclopropyl dienoic acid (3)28 is a potent RXR agonist. Novel aza-retinoids, exemplified by compound 4,29 as well as amide retinoids,30 have been described. RXR agonists based on aryl-trienoic acid compounds either unbranched31 or locked with one32 or multiple-fused33 ring systems were developed by Boehm and co-workers, of which 533 demonstrates the latter. Our group has reported that the addition of a single fluorine atom ortho to the carboxylic acid group of 1, giving compound 6,34 and the addition of two fluorine atoms, giving compound 7,35 increases RXR agonism in human colon cancer cell lines, Caco-2 and HCT-116, respectively. Incorporating a pyridine ring, compound 836 has been reported to be equally potent as 1, and substituting the alkene bridging group in 8 for a cyclopropyl bridging group yields LGD100268 (9),36 with a reported potency 1 order of magnitude greater than 1 in a CV-1 cell 8433

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

line.36 An unsaturated analogue of 1, compound 10,37 has a reported synthetic route that can provide preclinical trial quantities,38 although to our knowledge, the ability of 10 to bind and activate RXR has not been reported. Finally, acrylic acid 1139 demonstrates potent, selective RXR agonism, and acrylic acid CD2915 (12)40 is also a known RXR agonist.

Figure 1. Docking results for compounds 8−19 relative to 1. Calculated Autodock binding free energies were used to score the ligands, with negative values indicating better binding than 1. Circles, docking to 1MVC; diamonds, docking to 1H9U; triangles, docking to 3FUG; open symbols, using Babel charges; filled symbols, using Autodock charges.

Because of the potent and selective RXR agonism reported for compounds 8, 9, and 11 as well as a lack of reported biological evaluation of 10, recent work in our laboratories has focused on synthesizing seven novel analogues, 13−19, modeled on compounds 8−12 and subsequently assessing these novel compounds for RXR-selective activation alongside 1 and their parent compounds, 8−12.



RESULTS AND DISCUSSION Molecular Modeling. Docking results for compounds 8− 19 relative to 1 are given in Figure 1. Compounds were scored by the Autodock binding free energy estimate, with negative values indicating better binding than 1. Docks were performed with both the Autodock and OpenBabel charges; the latter tend to show less overpolarization.35 All compounds were predicted to bind nearly as well or better than 1, with predicted binding affinities generally within 0.5 kcal/mol of 1. Overlays of docking poses in the L-shaped binding pocket showed that the phenyl ring was held in nearly identical position for most ligands, while small adjustments were observed in the position of the double ring system. This is illustrated in Figure 2, which shows an overlay of the top docking binding poses of 1 and 13 in the binding pocket. The observation that chemical substitutions are accommodated by small shifts of the fused ring system indicates the presence of available space around the fused ring, which

Figure 2. Overlay of best poses for 1 (orange) and compound 13 (blue) in 1MVC using OpenBabel charges. Ile268, which makes important hydrophobic contacts, and was treated flexible in the docking studies, is shown as ball-and-stick.

suggests that the fused ring might be a good target for future chemical modifications. 8434

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Scheme 1

Scheme 2

8435

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Chemistry. The synthesis of pyrimidine-bexarotene analogue 13 and the pyrimidine-9 analogue 14 follows a scaled down methodology38 that was recently reported for the synthesis of pyridine-bexarotene analogues 8 and 9 (Scheme 1). We also undertook the synthesis of bexarotene analogue 8 and 9 to have standards against which to test 13 and 14. Commercially available dimethyl pyridine-2,5-dicarboxylate was selectively saponified by the addition of solid sodium hydroxide pellets to a suspension in methanol that was then refluxed for 4 h followed by acidification to give crude 5-(methoxycarbonyl)pyridine-2-carboxylic acid (20)38 in 74% after filtration and recrystallization from boiling water. Carboxylic acid 20 was converted to acid chloride 2138 by treatment with thionyl chloride, and acid chloride 21 was used in the aluminum chloride catalyzed Friedel−Crafts acylation reaction with compound 2234 to give ketone 2338 in 54% yield over two steps. Ketone 23 was treated with a 3.0 M solution of methyl magnesium chloride in THF at 0 °C, followed by quenching with a 1 N solution of hydrochloric acid and extraction, and the concentrated residue was refluxed with para-toluene sulfonic acid monohydrate to give alkene 2438 in 55% yield over three steps. Alkene 24 was converted to the cyclopropyl methyl ester 2538 in 82% yield by adding a solution of 24 in THF to a DMSO solution of trimethylsulfoxonium iodide treated with 22 wt % potassium tert-butoxide in THF. Both alkene 24 and cyclopropyl methyl ester 25 were saponified by treatment with aqueous potassium hydroxide and refluxing in methanol to give, after precipitation with 20% HCl, the corresponding carboxylic acids 8 and 9, respectively, in near quantitative crude yields. The carboxylic acids were purified by recrystallization from hot ethyl acetate and hexanes. Commercially available pyrimidine-2,5-dicarboxylic acid was refluxed in thionyl chloride with a few drops of catalytic DMF for 4 h, and excess thionyl chloride was removed in vacuo to give the solid diacid chloride that was dissolved in toluene and added to a solution of triethyl amine in methanol to yield the dimethyl pyrimidine-2,5-dicarboxylate 26 in 72% yield over two steps. A suspension of dimethyl ester 26 in methanol was then treated with sodium hydroxide pellets and refluxed for 4 h, followed by acidification with 2N HCl, to give 5-(methoxycarbonyl) pyrimidine-2,5-dicarboxylate (27) in 89% yield after filtration. Acid 27 was converted to acid chloride 28 by treatment with thionyl chloride, and acid chloride 28 was used in the aluminum chloride catalyzed Friedel−Crafts acylation reaction with compound 22 to give ketone 29 in 68% yield over two steps. Ketone 29 was treated with a 3.0 M solution of methyl magnesium chloride in THF at 0 °C followed by quenching with a 1 N solution of hydrochloric acid and then extracted, and the concentrated residue was refluxed with paratoluene sulfonic acid monohydrate to give alkene 30 in 40% yield over three steps. Alkene 30 was converted to the cyclopropyl methyl ester 31 in 68% yield by adding a solution of 30 in THF to a DMSO solution of trimethylsulfoxonium iodide treated with 22 wt % potassium tert-butoxide in THF. Both alkene 30 and cyclopropyl methyl ester 31 were saponified by treatment with aqueous potassium hydroxide and refluxing in methanol to give, after precipitation with 20% HCl, the corresponding carboxylic acids 13 and 14, respectively, in near quantitative crude yields. The carboxylic acids were purified by recrystallization from hot ethyl acetate and hexanes (Scheme 2).

Fortunately, compound 14 yielded single, transparent crystals from ethyl acetate, suitable for X-ray diffraction, and an X-ray diffraction study was performed (Figure 3).

Figure 3. X-ray crystal structure of 14. The thermal ellipsoid plot is shown at the 50% probability level.

To synthesize a sample of compound 10, a modified literature procedure37 was used. Toluene was reacted with dihydro-2,2,5,5-tetramethylfuran-3(2H)-one (32) with aluminum chloride as a catalyst to provide 3,4-dihydro-1,1,4,4,7pentamethylnaphthalen-2(1H)-one (33)37 in 44% yield. Ketone 33 was reacted with bromine and an aluminum chloride catalyst to afford 6-bromo-3,4-dihydro-1,1,4,4,7pentamethylnaphthalen-2(1H)-one (34). Ketone 34 was condensed with para-toluenesulfonyl hydrazide to give compound 35 in 50% yield. Compound 35 was treated with methyl lithium to provide 6-bromo-1,4-dihydro-1,1,4,4,7pentamethylnaphthalene (36)38 in a Shapiro reaction41 in 90% yield (Scheme 3). With alkene 36 in hand, we proceeded to make the appropriate coupling partner for it en route to compound 10 by reacting 4-cyanobenzoyl chloride (37) with N,O-hydroxylamine hydrochloride to give the Weinreb amide (38)38 in 88% yield (Scheme 4). To complete the synthesis of compound 10, we treated alkene 36 with butyl lithium followed by amide 38 to give ketone 3938 after aqueous acidic workup in 82% overall yield. The nitrile group of ketone 39 was hydrolyzed with potassium hydroxide to give carboxylic acid 4038 after aqueous acidic workup in 96% yield (Scheme 5). Acid 40 was treated with methyl magnesium chloride, followed by aqueous acidic workup to give alcohol 4138 that was subsequently refluxed with para-toluenesulfonic acid monohydrate in toluene to give alkene 10 in 91% overall yield (Scheme 5). To synthesize compound 15, a strategy using the Suzuki− Miyura coupling reaction42 as the key step was followed. Compound 22 was treated with bromine in DCM to give compound 4216b in 60% yield. Bromide 42 was treated with butyl lithium followed by triisopropylborate and aqueous acidic workup to give boronic acid 43 in 45% yield over three steps (Scheme 6). To prepare the Suzuki coupling partner for boronic acid 4338 en route to 15, 3-bromo-4-methylbenzoic acid (44) was treated with borane·THF to give a benzyl alcohol that was subsequently oxidized to aldehyde 4543 with manganese(IV)oxide in 74% overall yield. Aldehyde 45 was converted to the alkene ester 46 by a Horner−Wadsworth−Emmons reaction.44 With the alkene ester 46 in hand, we proceeded to couple it 8436

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Scheme 3

Scheme 4

Scheme 6

with boronic acid 43 via a Suzuki−Miyura reaction to give biphenyl 47 in 80% yield. The ester group of biphenyl 47 was saponified with KOH to give the carboxylic acid 15 in 75% yield after aqueous acidic workup (Scheme 7). Next, we targeted the synthesis of a trifluoro-methyl analogue of 11, compound 16, because we wished to compare the substitution of the hydroxyl group of 11 with a nonpolar methyl group (15) versus a much more polar trifluoro-methyl group (16). Hence, we performed the Horner−Wadsworth− Emmons reaction on commercially available aldehyde 48 to give ester 49 in 98% yield. With aromatic bromine 49 in hand, we proceeded to couple it with boronic acid 43 via a Suzuki− Miyura reaction to give biphenyl 50 in 71% yield. The ester group of biphenyl 50 was saponified with KOH to give the carboxylic acid 16 in 75% yield after aqueous acidic workup (Scheme 8). Next, we set out to synthesize an analogue of 15 with an alkene in the aliphatic ring: compound 17. Thus, we converted alkene 36 to boronic acid 5138 by treating 36 with butyl lithium followed by triisopropyl borate and an aqueous acidic workup. With boronic acid 51 in hand, we proceeded to couple it with ester 46 via a Suzuki−Miyura reaction to give biphenyl 52 in 72% yield. The ester group of biphenyl 52 was saponified with KOH to give the carboxylic acid 17 in 75% yield after aqueous acidic workup (Scheme 9).

To compare the effect of placing the alkene in the aliphatic ring of 11, we prepared compound 18. First, we made the appropriate aromatic halide coupling partner for boronic acid 51 by acetylating 3-bromo-4-hydroxybenzaldehyde (53) to give aldehyde 5439 in 90% yield, and then we carried out the Horner−Wadsworth−Emmons reaction on 54 to give ester 5539 in 84% yield. With aromatic bromide 55 in hand, we proceeded to couple it with boronic acid 51 via a Suzuki− Miyura reaction to give biphenyl 56 in 54% yield. The ester group of biphenyl 56 was saponified with KOH to give the carboxylic acid 18 in 89% yield after aqueous acidic workup (Scheme 10). Because of our recent finding34 that the addition of a fluorine atom ortho to the carboxylic acid group of 1 increases its binding and activation of RXR, we prepared 19 to investigate both the addition of an alkene in the aliphatic ring of 1 and fluorination ortho to the carboxylic acid. The synthesis of 19 begins with the sodium carbonate mediated conversion of methyl-4-acetyl-2-fluorobenzoate 57 to the enol triflate 58 in 27% yield. The enol triflate 58 was then coupled to 51 to give

Scheme 5

8437

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Scheme 7

human colon cancer (HCT-116) cells (Figure 4). This assay tests for ligand binding to a recombinant, full length RXR containing an activation domain. If the ligand−RXR complex then homodimerizes with an RXR−Gal4 fusion protein, the luciferase reporter gene will be transcribed, as the luciferase gene is downstream of Gal4p DNA binding elements. We tested the indicated compounds at two different concentrations (25 and 100 nM) in this system. The 100 nM assay (Figure 4B) revealed that five compounds (8, 9, 11, 13, and 15) exhibited high activity compared with compound 1 (bexarotene). Moreover, the 25 nM assay of compounds shows an even wider range of receptor binding and RXR homodimerization ability because the 100 nM assay likely approaches RXR binding saturation while the 25 nM assay represents a subsaturating concentration of ligand (see also EC50 values in Table 1). Compounds 8, 9, 11, 13, and 15 possess significantly (using a one-way ANOVA, p < 0.01) greater activity than compound 1 in the 25 nM assay (Figure 4A), with compound 18 also approaching statistical significance. Additionally, compounds 12, 17, and 19 display a smaller degree of RXR transcriptional response (using a oneway ANOVA, p < 0.01) relative to compound 1 in the 25 nM assay, and compounds 12 and 17 also possess significantly lower binding than compound 1 in the 100 nM assay, but 12, 17, and 19 still have high activity compared to the ethanol vehicle control (Figure 4A,B). These results imply that compounds modeled after 1 can be synthesized successfully and can mediate RXR transcriptional activation, and we are interested in elucidating the factors responsible for eliciting the different response-ranges observed. We have also utilized the mammalian two-hybrid assay in human HCT-116 cells to assess dose−response relationships (see Supporting Information Figures 1−3), and EC50 values as described below. Novel Analogues of 1 Bind to RXR and Mediate Transactivation in the Context of an RXRE. It is important to note that the use of the mammalian two-hybrid assay as an initial screen for agonist binding to and activation of RXR is useful because of the speed, convenience, and sensitivity of the assay. However, one potential drawback of the mammalian twohybrid assay is that the RXR−agonist complex may be dependent on the correct biologically relevant DNA platform, or retinoid X receptor responsive element (RXRE), that specifically associates with the RXR homodimer in vivo. The RXRE DNA sequence is usually found in the promoter region and also up- or downstream of the genes controlled by the RXR homodimer in response to the endogenous 9-cis RA ligand or when RXR is bound to a synthetic rexinoid. It is possible that

Scheme 8

Scheme 9

methyl ester 59 in 55% yield, and ester 59 was saponified with KOH in methanol to give 19 in 70% yield (Scheme 11). Finally, we prepared a sample of acrylic acid 12 to compare against 11 and the other novel acrylic acid analogues (15−18) in this study. Thus, 3-bromobenzaldehyde 60 was converted to ethyl acrylate 61 in 98% yield. The ethyl acrylate 61 was coupled to boronic acid 43 to give ethyl ester 62 in 70%, and 62 was subsequently saponified with KOH in methanol to yield 12 in 84% yield (Scheme 12). Biological Assays and Rationale. A Mammalian TwoHybrid Assay to Assess RXR Transcriptional Response of Analogues Compared to 1. Biological assessment of a subset of analogues described above (compounds 8−19) was first carried out employing the mammalian two-hybrid assay in 8438

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Scheme 10

low expression of RXRα47 and other receptors of interest to this study so we could quantitate most of the measurable effects as resulting from the expression of the transfected, rather than endogenous, receptor. The expression levels (as assessed by Western blotting) and ligand responsiveness of the HCT-116 cell line transfected with and without each of the receptors evaluated in this study are shown in Supporting Information Figure 4. We tested the indicated compounds at two different concentrations (25 and 100 nM) in the RXRE assay. The results in Figure 5 reveal that compounds 8, 9, 10, 11, 13, 15, and 18 displayed transcriptional activity consistently above the compound 1 level in both assays (Figure 5A,B). In particular, compounds 8, 11, 15, and 18 possess a significantly greater activity than compound 1 at both concentrations tested (using a one-way ANOVA, p < 0.05 for compound 8, 15, and 18, and p < 0.01 for compound 11). Importantly, the profile of analogue activity in the RXRE-based assays (Figure 5) is almost identical to the profile of analogues identified in the mammalian two-hybrid assay (Figure 4), thus validating the mammalian two-hybrid assay, and reinforcing the identification and reproducibility of those analogues with the greatest potency in terms of RXR activation. Evaluation of EC50 Values and Quantitation of RAR Agonist Activity. We utilized our mammalian two-hybrid assay in human colon cancer cells (Figure 4) to evaluate a much larger array of 1 and analogue concentrations. In addition to employing a single ligand concentration as in Figure 4, dose− response assays were carried out with ligand concentrations ranging from 10−9 M up to 0.3 × 10−5 M of each compound as well as 1 (Supporting Information Figures 1−3). Utilizing this collection of dose−response experiments, we were able to calculate EC50 values, which are listed in Table 1. Our EC50 value for 1, which is an estimate of ligand affinity for RXR, is similar to values obtained previously by another group.16b,17 In fact, Boehm and co-workers show that the different isoforms of RXR have similar EC50 values, within the same order of magnitude, not only for 1, but several analogues of 1.17 Moreover, the data in Figures 4 and 5 suggest that compounds 12, 16, 17, and 19 possess consistently lower RXR-mediated transcriptional activity while compounds 8, 9, 11, 13, 15, and 18 are consistently more active, observations that are entirely consistent with the EC50 values in Table 1. We also performed an analysis of the “residual” retinoic acid receptor (RAR) agonist activity of 1, as well as for each analogue versus the known RAR ligand (all-trans retinoic acid). The results of this

Scheme 11

Scheme 12

the RXRE may allosterically influence the affinity and/or selectivity of the RXR protein toward potential ligands. Thus, a second screening protocol for our collection of possible RXR agonists included transfection of HCT-116 cells with an expression vector for wild-type human RXRα along with a reporter construct that contains an authentic RXRE driving the expression of the luciferase reporter gene. The most frequent side effect of bexarotene administration is an increase in plasma triglycerides as a result of liver RXR/LXR-directed activation of lipid metabolism.45 Thus, we chose to first assess the ability of our analogues to bind/activate RXRα because the liver expresses predominantly the α isoform,46 and this isoform is likely responsible for the elevated triglyceride synthesis in the liver. We selected HCT-116 as a cell line that has a relatively 8439

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Figure 4. Identification of RXR agonists via a mammalian two-hybrid screening assay in human colon cancer cells. HCT-116 human colon cancer cells were cotransfected using a pCMV-hRXR binding domain vector (BD) as well as an hRXR-activation domain (AD) plasmid along with a pFRLuc reporter gene containing BD-binding sites and renilla control plasmid. Cells were transfected for 24 h utilizing a liposome-mediated transfection protocol and then treated with ethanol vehicle and either 25 or 100 nM of the indicated compound. After a 24 h incubation, cells were lysed and a dual luciferase assay was completed. Analogue-mediated RXR binding and homodimerization, as measured by luciferase output, was compared to the RXR agonist parent compound 1 (value set to 1.0). Values are means ± SD *p < 0.05, **p < 0.01 vs compound 1.

known apoptotic inducer (sodium butyrate, NaBu) serves as a positive control in this system. Evaluation of 1 and Analogues As Inhibitors of VDR−RXR Activity Due to Analogue-Mediated Formation of RXR−RXR Homodimers. 1 binds to RXR and induces RXR−RXR homodimerization, thus there may be less RXR available in the intracellular pool for the creation of heterodimers with other nuclear receptors such as the formation of VDR−RXR heterodimers. This disruption of RXR heterodimer pathways is one of several potential molecular mechanisms that may be responsible for side effects of 1 in humans. Therefore, we seek to develop RXR analogues that are not only more robust in their RXR biopotency via RXRE-mediated transcription but also that are less disruptive to RXR heterodimerization pathways. As a result, we measured vitamin D responsive element (VDRE)-mediated transcriptional activity in the presence of the active 1,25-dihydroxyvitamin D3 metabolite (1,25D; 10−9 M) to assess the level of RXR−RXR “diversion” and thus likely inhibition of VDR−RXR-directed transcription at two different concentrations of RXR analogues (Figure 7A, rexinoid 10−7 M; Figure 7B, rexinoid 10−6 M). Compound 1 inhibited VDR activity by approximately 50% in the 10−6 M assay (Figure 7B) and by 30% in the 10−7 M assay (Figure 7A). Compounds 8, 10, 13, 14, and 17 inhibited VDR activity less compared with compound 1 in both assays (Figure 7A,B). Statistically, compounds 1, 9, 11, and 15 are significantly lower (using a one-way ANOVA, p < 0.05 for compounds 1, p < 0.01 for compounds 9, 11, and 15) than 1,25D alone in the 100 nM assay (Figure 7A). Most compounds (except for 10) are significantly lower (using a one-way ANOVA, p < 0.05 for compounds 17, p < 0.01 for compounds 1, 8, 9, 11, 12, 13, 14, 15, 16, 18, and 19) than 1,25D alone in the 1000 nM assay (Figure 7B). These results suggest that the expected side effects of compound 8, 10, 13, 14, and 17 due to RXR−RXR diversion may be smaller than compound 1. It is important to note that some previous reports50 have demonstrated a stimulatory effect

assay, which employed expression of human RARα (the most widely distributed isoform of RAR in human tissues) and a retinoic acid responsive element (RARE)-luciferase reporter system, revealed that compounds 8, 10, 13, 14, 15, 17, and 19 possess greater RAR agonist activity, compounds 9 and 18 are approximately equal to 1 in its activation of RAR, and compounds 11, 12, and 16 possess slightly lower RAR-directed activity. Taken together, these results suggest that modification of 1 with more polar atoms in the aromatic ring that bears the carboxylic acid may not only increase the analogues’ ability to activate RAR (compounds 8, 13, 14, 19), but also increase its ability to activate RXR (analogues 8 and 13). Analysis of Apoptotic Activity by Bexarotene and Novel Analogues in a CTCL System. One potential mechanism to explain the efficacy of 1 in treating CTCL effectively is that this rexinoid induces apoptosis and/or cytotoxicity in the Tlymphocyte.48 Thus, we next tested CTCL (Hut-78) cells treated with 1 and the indicated analogues for their ability to induce classic apoptosis, using an assay for caspases 3 and 7, two caspases that mediate apoptosis.49 We tested each compound (8−19) by treatment for either 24 or 48 h (Figure 6). The results of the apoptosis assay suggest that most of the analogues possess some pro-apoptotic activity. Two compounds (analogues 8 and 13) that exhibit higher RXR binding affinity (lower EC50, Table 1) and greater potency in the RXR transcription assays (Figures 4 and 5) are also more active (Figure 6) than compound 1 in the apoptotic assay (using a one-way ANOVA, p < 0.05 for compound 8, p < 0.01 for compound 13 both at 48 h, and p < 0.05 for compound 13 at 24 h). Several analogues reveal activity that is greater than the ethanol vehicle control (Figure 6) in eliciting apoptosis at 24 h (using a one-way ANOVA, p < 0.05 for compounds 1, 9, 10, 12, 14, and 18, p < 0.01 for compounds 8, 13, 16, 17, and 19) or 48 h (using a one-way ANOVA, p < 0.05 for compounds 1, 9, 14, 17, 18, and 19, p < 0.01 for compounds 8, 13). A potent 8440

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Table 1. Determination of EC50 Values in RXR M2H Assay and Quantitation of RAR Agonist Activitya

(1) EC50 values were determined from full dose−response experiments with each compound in the range of 10−9 to 10−5 M in transfected HCT116 cells using an RXR mammalian two-hybrid system. (2) RAR agonist activity was derived from an RAR/RARE reporter system in transfected HCT-116 cells treated with all-trans RA or indicated analogue at 100 nM or 1 μM. The activity with analogue (or compound 1) divided by the activity with all-trans RA expressed as a percentage represents the RAR agonist activity. a

conducted additional transcriptional assays with VDR−RXR employing different classes of vitamin D responsive elements (VDREs) beyond the ER6 VDRE used in Figure 7A,B. These experiments (Figure 7C−F) reveal that the effect of rexinoids (1/analogues) on 1,25D−VDR−RXR transcriptional response

of 9-cis RA/rexinoids on 1,25D−VDR−VDRE-mediated transcription. However, several other studies have revealed the opposite effect, i.e., suppression of VDR−RXR activity (see Introduction). Therefore, this remains a controversial theme in the vitamin D field. To address this important question, we 8441

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Figure 5. Evaluation of RXR agonists via an RXRE-luciferase reporter-based assay in human colon cancer cells. HCT-116 cells were transfected with human RXRα, RXRE-luciferase reporter gene, and renilla control plasmid. Cells were transfected for 24 h utilizing a liposome-mediated transfection protocol as in Figure 4 and then treated with ethanol vehicle and 25 or 100 nM of the indicated compound. After a 24 h incubation, cells were lysed and a luciferase assay was completed. Analogue-stimulated, RXR-mediated transcription, as measured by luciferase output, was compared to the RXR agonist parent compound 1 (value set to 1.0). Values are means ± SD *p < 0.05, **p < 0.01 vs compound 1.

Figure 6. Assessment of bexarotene and analogues for apoptotic activity in CTCL cells. Human cutaneous T lymphocytes (Hut-78) were plated and subsequently dosed with the indicated treatments. Cells were allowed to incubate for 24 or 48 h, followed by measurement of apoptosis with a commercial kit (see Experimental Section). Sodium butyrate (NaBu), a known inducer of apoptosis in this system, was used as a positive control. Values are means ± SD *p < 0.05, **p < 0.01 vs compound 1. #p < 0.05, ##p < 0.01 vs ethanol.

contains 2 VDREs, has a net zero effect of most rexinoids on 1,25D−VDR transactivation (Figure 7E,F) with only a few analogues showing significant inhibiton (using a one-way ANOVA, p < 0.05 for compounds 18 and 19, p < 0.01 for compounds 12 in the 100 nM assay, p < 0.05 for compounds 12 and 18 in the 1000 nM assay). Related findings have been reported for a TR−RXRβ−TRE system where RXRβ can either enhance, not alter, or inhibit TR-dependent transcription depending on the nature of TRE.51 These novel results with VDR not only address the need to evaluate the effect of 1/ rexinoids on 1,25D−VDR−RXR transcriptional activity in the context of multiple VDREs, but they may also provide a compelling resolution to the conflicting data that have been reported in the literature with respect to the actions of rexinoids on vitamin D signaling. While much of the comparative discussion has focused on how a given novel analogue (13−19) performs in these assays

(e.g., repression, no effect, stimulation) is dependent on the nature of the VDRE. For example, as shown in Figure 7A,B, there is a reproducible inhibition with 1 and most analogues on VDR activity (as described above) when the VDR DNA platform is an everted repeat-6 (ER6) from the CYP3A4 gene. In contrast, there is a stimulation in VDR-mediated activity when using the more classical direct repeat-3 (DR3) VDRE (Figure 7C,D) in the presence of 1,25D and some of the analogues compared to treatment with 1,25D alone. Compounds 1 and 8 are significantly higher (using a one-way ANOVA, p < 0.05) than 1,25D alone, and compound 12 and 17 are significantly lower (using a one-way ANOVA, p < 0.05) than 1,25D alone in the 100 nM assay (Figure 7C). Additionally, compounds 1, 8, and 15 are significantly higher (using a one-way ANOVA, p < 0.05 for compounds 8 and 15, p < 0.01 for compounds 1) than 1,25D alone in the 1000 nM assay (Figure 7D). The CYP24 natural promoter, which 8442

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Figure 7. Evaluation of bexarotene and selected analogues for modulation of VDR−VDRE-mediated transcription via formation and/or diversion of RXR homodimers. HCT-116 cells were transfected with hVDR, VDRE-luciferase reporter plasmids (proximal ER6 or distal DR3 from the human CYP3A4 gene or the natural human CYP24A1 promoter-5.5kb), and a renilla control plasmid. Cells were transfected for 24 h utilizing a liposomemediated transfection protocol and then treated with ethanol vehicle, 1 nM 1,25D, and 100 or 1000 nM of the indicated compound in the presence of 1 nM 1,25D. After a 24 h incubation, cells were lysed and a luciferase assay was completed. 1,25D-stimulated, VDRE-mediated transcription in the presence of rexinoids (as measured by luciferase output) was compared to the 1,25D-only group (value set to 1.0). (A) ER6-mediated activity in the presence of 1 nM 1,25D and 100 nM analogues. (B) ER6-mediated activity in the presence of 1 nM 1,25D and 1000 nM analogues. (C) DR3mediated activity in the presence of 1 nM 1,25D and 100 nM analogues. (D) DR3-mediated activity in the presence of 1 nM 1,25D and 1000 nM analogues. (E) hCYP24A1 promoter activity in the presence of 1 nM 1,25D and 100 nM analogues. (F) hCYP24A1 promoter activity in the presence of 1 nM 1,25D and 1000 nM analogues. Values are means ± SD *p < 0.05, **p < 0.01 vs 1 nM 1,25D only (black bar).

of 11 to give analogue 18 conferred similar RXRα EC50 values and RAR agonist activities between 18 and 11 as well as similar apoptosis and VDRE transcription levels. However, substituting a methyl group for the hydroxyl group in 11 while simultaneously introducing an unsaturation in the aliphatic ring to give analogue 17 resulted in a higher RXRα EC50 value as well as higher RAR agonist activities than observed in 11. Bexarotene and the Novel Analogues Are Not Mutagenic. Utilizing the EPA-approved Saccharomyces cerevisiae mutagenesis assay,34 all compounds were tested for their ability to damage DNA. DNA damage in this assay leads to either a point mutation (reversion of a phenotype of isoleucine auxotroph to isoleucine prototroph) or substantial damage leading to subsequent repair. In this way, DNA mutagenesis in this strain can be quantified by colony formation on selective media, indicative of a phenotype change induced by DNA damage. All compounds were tested at concentrations up to 0.15% w/v and

versus 1, it is also useful to examine how each novel analogue performs relative to a known compound (8−12) possessing a correspondingly more similar structure. Substituting a pyrmidine ring in 13 and 14 versus a pyridine ring in 8 and 9 resulted in higher RXRα EC50 values, whereas the RAR agonist activity was similar, as was the ability to modulate VDR activity on ER6 and CYP24, and the ability of 13 and 14 to stimulate apoptosis. On the other hand, the fluorine substitution in novel analogue 19 versus the hydrogen atom in 10 resulted in similar RXRα EC50 values, whereas the RAR agonist activity for 19 was much greater than 10, but the apoptosis and ER6/CYP24 VDRE transcription levels were similar for both 10 and 19. Substituting a methyl group, 15, or trifluoromethyl group, 16, for the hydroxyl group of 11 led to higher RXRα EC50 values, whereas the RAR agonist activity was higher in 15 but nearly identical in 16. The apoptosis profile and activity on all three VDREs were similar for 11, 15, and 16. Interestingly, introducing an unsaturation in the aliphatic ring 8443

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

ng of the renilla control plasmid and 2 μL/well of Express-IN was again used for the liposome-mediated delivery. The cells were incubated for 24 h post-transfection and then treated with media containing ethanol vehicle (0.1%), or analogues (final concentration of 100 nM or 1 μm). After a 24 h incubation period, the amount of retinoid activity was measured using the same luciferase assay described above. Three independent assays were conducted with triplicate samples for each treatment group. EC50 Determination. EC50 values were determined from full dose−response curves ranging from 1 × 10−9 to 0.3 × 10−5 M in transfected HCT-116 cells using an RXR mammalian two-hybrid system. HCT-116 cells were plated overnight at 80000 cells/well in a 24-well plate and maintained as described above. The cells were cotransfected using a human RXR binding domain (BD) vector, a human RXR activation domain (AD) vector, a luciferase reporter gene containing BD-binding sites, and renilla control plasmid. Transfection was achieved via 2 μL/well of Express-IN transfection reagent which was allowed to incubate for 24 h with the cells. Then, the cells were treated with ethanol vehicle (0.1%) or analogues (1.0, 2.5, 5.0, 7.5, 10, 25, 50, 75, 100, 250, 500 nM, 1, 2, 3 μM) and incubated for 24 h. The amount of rexinoid activity at each concentration was measured using the same luciferase assay described above, and EC50 values were derived from dose−response curves of ligand concentration versus normalized luciferase activity RARE-Mediated Transcription Assay. The RARE assay was completed using HCT-116 cells plated at 80000 cells/well in a 24-well plate and maintained as described above. The cells were cotransfected using 250 ng of pTK-DR5(x2)-luciferase reporter gene. This RARE is an optimized element that has been described previously54 and is responsive to the RAR ligand, all-trans retinoic acid. Also included in the transfection were 50 ng of pCMX-human RARα and 20 ng of the renilla control plasmid, along with 2 μL/well of Express-IN for liposome-mediated delivery of the DNA into the cells. The cells were incubated for 24 h post-transfection and then treated with ethanol (0.1%) or analogues (final concentration of 100 nM or 1 μM). After a 24 h incubation period, the amount of retinoid activity was measured using the same luciferase assay described above. The activity with analogue (or compound 1) divided by the activity with all-trans RA (expressed as a percentage) represents the RAR agonist activity. Three independent assays were conducted with triplicate samples for each treatment group. Apoptosis Assay. Apoptotic activity was assessed by the CaspaseGlo 3/7 assay (Promega, Madison, WI) according to the manufacturer’s instructions. The Caspase-Glo 3/7 assay is based on the cleavage of the DEVD sequence of a luminogenic substrate by caspases 3 and 7, which results in a luminescent signal. HuT-78 (human T-cell lymphoma) cells were distributed (1 × 104 cells/well) in white-walled 96-well microplates (Corning, NY) in 100 μL of medium and incubated with 500 μM sodium butyrate (NaBu), 100 nM 1, or rexinoid analogues for 24 and 48 h. The Caspase-Glo 3/7 reagent was then added to each well and incubated for an additional 1 h at room temperature. The luminescence was measured in a luminometer (Safire2, Tecan, US). NaBu, a known inducer of apoptosis in HuT-78, was used as a positive control. Each treatment group was dosed in triplicate, and at least two independent experiments were performed. Numbers were standardized to sodium butyrate (set at 100%). VDR−VDRE Assay. The VDR−VDRE assay was completed using HCT-116 cells plated at 80000 cells/well in a 24-well plate and maintained as described above. The cells were cotransfected using 250 ng of the indicated VDRE-luciferase reporter gene (proximal ER6 or distal DR3 VDRE from the human CYP3A4 gene, or the natural human CYP24A1 promoter-5.5kb), 50 ng of pSG5-hVDR, and 20 ng of the renilla control plasmid with 2 μL/well of Express-IN used for liposome-mediated DNA delivery. The cells were incubated for 24 h post-transfection and then further treated with ethanol vehicle (0.1%), 10−9 M 1,25-dihydroxyvitamin D3 (1,25D) alone, or 10−9 M 1,25D in combination with analogues (final concentration of 10−6 M or 10−7 M). After a 24 h incubation period, the amount of luciferase activity was measured using the luminescence assay described above. Three

none increased colony formation above vehicle control (data not shown).



CONCLUSIONS 1 is an FDA approved drug to treat cutaneous T-cell lymphoma and is being explored to treat a myriad of additional types of cancer. Moreover, recent data suggest that 1 is also a promising compound in the continued fight against AD.27,52 Our work was motivated by the untoward side effects of 1 because the drug appears to be safe and efficacious in treatment of some CTCL patients, but the side effects include hyperlipidemia and hypothyroidism due to the ability of 1 to serve as an RXR ligand with little heterodimer-specific selectivity. The potential benefit of exploring additional RXR ligands for therapeutic treatments is further motivated by the opportunities of rational drug design, and as we have shown,34,35 modeling can help drive synthesis of additional potent RXR ligands. Our continued work has produced seven novel compounds, four of which possess EC50 values lower than 1 and two of which possess superior pro-apoptotic profiles in CTCL cells. We are thus hopeful that an RXR ligand with a lower EC50 will translate into a pharmaceutical that would allow for a lower effective dose regime. Furthermore, side effects of hyperlipidemia and hypothyroidism are an ongoing consideration in treatment with 1. We address concerns about cross-talk by RXR ligands with other RXR nuclear receptor pathways, as well as the potential for new rexinoids to titrate away RXR from nonpermissive heteropartners by assessing our novel compounds for their ability to modulate VDR−RXR-directed transcription utilizing multiple VDREs. We demonstrate that many of our novel analogues are superior at mitigating RXR heterodimerization disruption compared to 1. Taken together, our novel RXR ligands appear more potent, demonstrate higher efficacy, and are less likely to lead to side effects than the parent compound. Work is ongoing to analyze further biological parameters of these compounds and develop additional novel analogues of 1 with improved side effect profiles.



EXPERIMENTAL SECTION

Mammalian Two-Hybrid Assay. HCT-116 colorectal carcinoma cells were plated overnight at 80000 cells/well in a 24-well plate and maintained in DMEM/high glucose (Hyclone) enhanced with 10% FBS (Invitrogen), 1 mM sodium pyruvate (Invitrogen), 100 μg/mL streptomycin, and 100 unit/mL penicillin. The cells were cotransfected using a human RXR binding domain (BD) vector, a human RXR activation domain (AD) vector, a luciferase reporter gene containing BD-binding sites, and renilla control plasmid. A liposome-mediated transfection was completed according to the manufacturer’s protocol using 2 μL/well of Express-IN transfection reagent (Thermo Fisher Scientific, Lafayette, CO) and allowed to incubate for 24 h. The cells were then treated with media containing ethanol vehicle (0.1%), bexarotene or analogues at a final concentration of 25 nM or 100 nM and incubated for 24 h. The amount of rexinoid activity was measured by luciferase output utilizing a dual-luciferase reporter assay system according to the manufacturer’s protocol (Promega, Madison, WI) in a Sirus luminometer (Berthold Detection System, Pforzheim, Germany). Three independent assays were conducted with triplicate samples for each treatment group. RXRE-Mediated Transcription Assay. The RXRE assays were completed using HCT-116 cells plated at 80000 cells/well in a 24-well plate and maintained as described above. The cells were cotransfected using 250 ng of RXRE-luciferase reporter gene (RXRE from the naturally occurring responsive element in the rat cellular retinol binding protein II gene: 53 AAAATGAACTGTGACCTGTGACCTGTGACCTGTGAC), 50 ng of pSG5-human RXRα, and 20 8444

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

independent assays were conducted with triplicate samples for each treatment group. Modulation of vitamin D activity was determined by evaluation of 1,25D-treated cells compared to cells treated with 1,25D plus bexarotene/analogue. Mutagenicity Assays. Mutagenicity assays utilized the D7 strain of Saccharomyces cerevisiae.55 D7 cells were incubated at 30 °C with shaking for 3 h with the compound solubilized in DMSO in increasing concentrations up to 0.15% w/v, and then aliquots were plated on selective media as described in Marshall56 and allowed to incubate for 4 days. Colony number was then scored and compared to DMSO alone (negative) and ethidium bromide (positive) controls. Docking Studies. Docking studies on the human RXRα (Protein Data Bank codes 1MVC57 and 3FUG,39 respectively) and RXRβ (Protein Data Bank code 1H9U58) were performed using the Autodock 4.2 software.59 As in our previous studies,34 protein residues Arg316 and Ile268 (residue numbering as in 1MVC) were treated flexibly and both Autodock and OpenBabel 2.3.0 charges60 were used. Docking used the Lamarckian genetic algorithm with 25 000 000 energy evaluations per dock and a total of 250 docks per compound. Calculated binding free energies were used to score the compounds. Instrumentation. A 400 MHz Bruker spectrometer was used to acquire 1H NMR and 13C NMR spectra. Chemical shifts (δ) are listed in ppm against residual nondeuterated solvent peaks in a given deuterated solvent (e.g., CHCl3 in CDCl3) as an internal reference. Coupling constants (J) are reported in Hz, and the abbreviations for splitting include: s, single; d, doublet; t, triplet; q, quartet; p, pentet; m, multiplet; br, broad. All 13C NMR spectra were acquired on a Bruker instrument at 100.6 MHz. Chemical shifts (δ) are listed in ppm against deuterated solvent carbon peaks as an internal reference. High resolution mass spectra were recorded using either a JEOL GCmate (2004), a JEOL LCmate (2002) high resolution mass spectrometer, or an ABI Mariner (1999) ESI-TOF mass spectrometer. General Procedures. Removal of volatile solvents transpired under reduced pressure using a Büchi rotary evaporator and is referred to as removing solvents in vacuo. Thin layer chromatography was conducted on precoated (0.25 mm thickness) silica gel plates with 60F-254 indicator (Merck). Column chromatography was conducted using 230−400 mesh silica gel (E. Merck reagent silica gel 60). All tested compounds were analyzed for purity by combustion analysis through Columbia Analytical Services (formerly Desert Analytics in Tucson, AZ) and were found to be >95% pure. 5-(Methoxycarbonyl)pyridine-2-carboxylic Acid (20). The protocol of Faul et al. was followed.38 To a suspension of dimethyl pydrine-2,5-dicarboxylate (10.09 g, 51.7 mmol) in methanol (130 mL) was added sodium hydroxide pellets (2.20 g, 55.0 mmol), and the heterogeneous reaction was stirred and refluxed for 4 h. After cooling to 65 °C, 2.0 N HCl (38 mL, 76 mmol) was slowly added, and over the course of addition, the solid dissolved and a precipitate formed. The reaction solution was allowed to cool to room temperature, and then it was cooled in an ice-bath and filtered. The filter-cake was washed with water to give an off-white product (7.71 g, 82%). A sample of this crude, filtered product (1.09 g) was dissolved in boiling water (70 mL), and the solution was slowly cooled to room temperature and then cooled in an ice-bath before it was filtered to give a white, crystalline powder (0.98 g) at a 74% overall yield, mp 189−190 °C. 1H NMR (400 MHz, DMSO-d6) δ 9.15 (dd, J = 2.0, 0.8, 1H), 8.44 (dd, J = 8.0, 2.0, 1H), 8.15 (dd, J = 8.0, 0.8, 1H), 3.91 (s, 3H), 3.34 (br s, 1H). 13C NMR (100.6 MHz, DMSO-d6) δ 165.5, 164.6, 151.7, 149.8, 138.3, 127.9, 124.6. IR (neat) ν 3440, 2969, 1717, 1694 cm−1. LC-FAB-MS (M + H)+ calcd for C8H8NO4 182.0453, found 182.0458. Methyl 6-[(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydronaphthalen-2-yl)carbonyl]nicotinate (23). The procedure of Faul and co-workers was followed.38 Methyl 6-(chlorocarbonyl)-pyridine-3carboxylate (21) was synthesized by refluxing 5-(methoxycarbonyl)pyridine-2-carboxylic acid (20) (1.20 g, 6.62 mmol) in thionyl chloride (12.0 mL, 165 mmol) in a 100 mL one-neck round-bottom flask fitted with a water-cooled reflux condenser. Excess thionyl chloride was removed in vacuo to give crude 21 as an off-white solid, and this solid was dissolved in dry benzene (ca. 20 mL) and evaporated to dryness to

remove residual thionyl chloride. The acid chloride 21 was dried on high vacuum to remove residual benzene. To a two-neck, 50 mL round-bottom flask equipped with a reflux condenser and magnetic stir-bar was added 22 (1.40 g, 6.91 mmol) followed by a solution of crude acid chloride 21 (6.62 mmol) in DCM (15 mL). Aluminum chloride (2.20 g, 16.5 mmol) was added to the reaction solution at room temperature slowly, with stirring, and the reaction solution turned from colorless to red accompanied by the evolution of gas and heat. The reaction was stirred for 5 min then heated to reflux for 15 min. The reaction was judged to be complete by TLC, and the solution was poured into an ice solution (25 mL) acidified with a 20% HCl solution (8 mL), and ethyl acetate was added (13 mL). The aqueous and organic layers were separated, and the aqueous layer was extracted with ethyl acetate (15 mL, twice). The combined organics were washed with water and brine, dried over sodium sulfate, filtered, and concentrated to give crude 23. Crude 23 was purified by column chromatography (250 mL SiO2, hexanes:ethyl acetate 95:5 to 85:15) to give 23 (1.30 g, 54%) as an off-white, crystalline solid, mp 122−125 °C. 1H NMR (400 MHz, CDCl3) δ 9.26 (dd, J = 2.0, 0.8, 1H), 8.49 (dd, J = 8.0, 2.0, 1H), 8.10 (dd, J = 8.0, 0.8, 1H), 7.42 (s, 1H), 7.21 (s, 1H), 3.99 (s, 3H), 2.39 (s, 3H), 1.68 (s, 4H), 1.29 (s, 6H), 1.20 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 195.8, 165.1, 158.6, 149.9, 149.2, 141.6, 138.1, 135.7, 133.2, 130.3, 129.7, 127.5, 123.8, 52.7, 34.8, 34.8, 34.4, 33.8, 31.6, 31.5, 20.5. IR (neat) 2919, 1720, 1688 cm−1. LCMS (M + H)+ calcd for C23H28NO3 366.2069, found 366.2070. Methyl 6-[(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydronaphthalen-2-yl)ethenyl]nicotinate (24). The procedure of Faul and co-workers was followed.38 To a 100 mL round-bottom flask charged with 23 (1.0077 g, 2.757 mmol) was added toluene (10 mL), and the solution was cooled in a salt-water ice bath to −15 °C with stirring, under nitrogen. To this solution was added a 22 wt % solution of MeMgCl in THF (1.20 mL, 3.60 mmol), and the reaction was stirred for 15 min at −15 °C and then warmed to room temperature and stirred for 35 min before quenching with 1N HCl (7 mL, 7 mmol). The reaction mixture was then separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were filtered and a small yellow filter-cake was dissolved in chloroform and added to the organic filtrate, and the combined organic filtrate was concentrated in vacuo to give a crude alcohol intermediate that was used without further purification. To this crude intermediate in a 100 mL roundbottom flask was added p-toluenesulfonic acid monohydrate (0.5247 g, 2.76 mmol) and toluene (40 mL), and the reaction was refluxed for 3 h into a Dean−Stark apparatus half-filled with toluene (6 mL). After the reaction had cooled to room temperature, it was added to a solution of sodium carbonate (0.78 g) in water (15 mL), shaken vigorously, and the layers were separated. The aqueous layer was extracted with ethyl acetate, and the combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude brown product that was purified by column chromatography (150 mL SiO2, hexanes:ethyl acetate 97.5:2.5 to 95:5) to give 24 (0.5595 g, 55%) as a white, fiberlike solid, mp 166−167 °C. 1H NMR (400 MHz, CDCl3) δ 9.22 (dd, J = 2.0, 0.8, 1H), 8.15 (dd, J = 8.0, 2.0, 1H), 7.14 (s, 1H), 7.11 (s, 1H), 7.02 (dd, J = 8.4, 0.8, 1H), 6.54 (d, J = 2.0, 1H), 5.51 (d, J = 2.0, 1H), 3.94 (s, 3H), 1.98 (s, 3H), 1.69 (s, 4H), 1.31 (s, 6H), 1.26 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 165.8, 161.1, 150.6, 148.0, 144.5, 142.5, 137.6, 136.7, 132.7, 128.0, 124.1, 121.1, 121.0, 52.2, 35.1, 35.0, 33.9, 33.8, 31.9, 31.8, 19.8. IR (neat) 2959, 1722, 1591 cm−1. LC-MS (M + H)+ calcd for C24H30NO2 364.2277, found 364.2272. Methyl 6-[(3,5,8,8-Pentamethyl-5.6,7,8-tetrahydronaphthalen-2-yl)cyclopropyl]nicotinate (25). The procedure of Faul and co-workers was followed.38 To a suspension of trimethylsulfoxonium iodide (0.365 g, 1.66 mmol) in DMSO (1.2 mL) was added a 20 wt % solution of potassium tert-butoxide in THF (0.94 mL, 1.67 mmol). A solution of 24 (0.40 g, 1.10 mmol) in THF (4.8 mL) was added dropwise over 20 min at 30−34 °C with stirring. The reaction was stirred for 60 min at 35 °C, then cooled to room temperature and quenched with 1N HCl (5 mL). The layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and 8445

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

5-(Methoxycarbonyl)pyrimidine-2-carboxylic Acid (27). To a suspension of of dimethyl pyrimidine-2,5-dicarboxylate (26) (3.34 g, 17.0 mmol) in methanol (30 mL) was added sodium hydroxide pellets (0.748 g, 18.7 mmol), and the heterogeneous reaction was stirred and refluxed for 4 h. After cooling to 65 °C, 2.0 N HCl (14 mL, 28 mmol) was slowly added. The reaction solution was allowed to cool to room temperature, and then it was concentrated in vacuo to give a solid. The solid was filtered with cold water, and the filter-cake was washed with a small amount of cold water to give 27 as a white crystalline solid (mp 152−154 °C) (2.765 g, 89%). 1H NMR (400 MHz, DMSO-d6) δ 13.89 (br s, 1H), 9.39 (s, 2H), 3.97 (s, 3H). 13C NMR (100.6 MHz, DMSO-d6) δ 164.3, 163.4, 159.6, 158.4, 124.8, 52.9. IR (neat) 3493, 2970, 1716, 1584, 1558 cm−1. GC-MS (M)+ calcd for C7H6N2O4 182.0328, found 182.0335. Methyl 2-[(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydronaphthalen-2-yl)carbonyl]pyrimidine-5-carboxylate (29). Methyl 2(chlorocarbonyl)-pyrimidine-5-carboxylate (28) was synthesized by refluxing 5-(methoxycarbonyl)pyrimidine-2-carboxylic acid (27) (1.22 g, 6.70 mmol) in thionyl chloride (12.0 mL, 165 mmol) in a 100 mL one-neck round-bottom flask fitted with a water-cooled reflux condenser. Excess thionyl chloride was removed in vacuo to give crude 28 as an off-white solid, and this solid was dissolved in dry benzene (ca. 20 mL) and evaporated to dryness to remove residual thionyl chloride. The acid chloride 28 was dried on high vacuum to remove residual benzene. To a two-neck, 50 mL round-bottom flask equipped with a reflux condenser and magnetic stir-bar was added 22 (1.45 g, 7.16 mmol) followed by a solution of crude acid chloride 28 (6.70 mmol) in DCM (15 mL). Aluminum chloride (2.20 g, 16.5 mmol) was added to the reaction solution at room temperature slowly, with stirring, and the reaction solution turned from colorless to red accompanied by the evolution of gas and heat. The reaction was stirred for 5 min then heated to reflux for 15 min. The reaction was judged to be complete by TLC, and the solution was poured into an ice solution (25 mL), acidified with a 20% HCl solution (8 mL), and ethyl acetate added (13 mL). The aqueous and organic layers were separated, and the aqueous layer was extracted with ethyl acetate (15 mL, twice). The combined organics were washed with water and brine, dried over sodium sulfate, filtered, and concentrated to give crude 29. Crude 29 was purified by column chromatography (250 mL SiO2, hexanes:ethyl acetate 95:5 to 85:15) to give 29 (1.677 g, 68%) as an off-white, crystalline solid, mp 94−96 °C. 1H NMR (400 MHz, CDCl3) δ 9.42 (s, 2H), 7.40 (s, 1H), 7.21 (s, 1H), 4.03 (s, 3H), 2.43 (s, 3H), 1.66 (s, 4H), 1.28 (s, 6H), 1.17 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 192.8, 165.9, 163.5, 158.4, 150.3, 142.0, 136.7, 131.9, 130.8, 130.0, 124.0, 52.9, 34.7, 34.6, 34.4, 33.8, 31.6, 31.4, 20.9. IR (neat) 2951, 1715, 1683, 1577, 1549 cm−1. LC-MS (M + H)+ calcd for C22H27N2O3 367.2022, found 367.2017. Methyl 2-[(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydronaphthalen-2-yl)ethenyl]pyrimidine-5-carboxylate (30). To a 100 mL round-bottom flask charged with 29 (1.0373 g, 2.83 mmol) was added toluene (10 mL), and the solution was cooled in a salt-water ice bath to −15 °C with stirring, under nitrogen. To this solution was added a 22 wt % solution of MeMgCl in THF (1.20 mL, 3.60 mmol), and the reaction was stirred for 15 min at −15 °C and then warmed to room temperature and stirred for 35 min before quenching with 1N HCl (7 mL, 7 mmol). The reaction mixture was then separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were filtered and a small yellow filter-cake was dissolved in chloroform and added to the organic filtrate and the combined organic filtrate and combined organic filtrate was concentrated in vacuo to give a crude alcohol intermediate that was used without further purification. To this crude intermediate in a 100 mL roundbottom flask was added p-toluenesulfonic acid monohydrate (0.5247 g, 2.76 mmol) and toluene (40 mL), and the reaction was refluxed for 3 h into a Dean−Stark apparatus half-filled with toluene (6 mL). After the reaction had cooled to room temperature, it was added to a solution of sodium carbonate (0.78 g) in water (15 mL), shaken vigorously, and the layers separated. The aqueous layer was extracted with ethyl acetate, and the combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to

concentrated to give an off-white crude solid that was purified by column chromatography (150 mL SiO2, hexanes:ethyl acetate 97.5:2.5 to 90:10) to give 25 (0.3411 g, 82%) as a white crystalline solid, mp 168−170 °C. 1H NMR (400 MHz, CDCl3) δ 9.08 (dd, J = 2.4, 0.8, 1H), 7.98 (dd, J = 8.4, 2.4, 1H), 7.27 (s, 1H), 7.11 (s, 1H), 6.74 (dd, J = 8.4, 0.8, 1H), 3.90 (s, 3H), 2.11 (s, 3H), 1.82−1.83 (m, 2H), 1.69 (s, 4H), 1.35−1.36 (m, 2H), 1.30 (s, 6H), 1.27 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 169.2, 166.1, 150.4, 143.8, 142.6, 137.1, 136.5, 135.7, 129.2, 128.3, 122.1, 120.7, 52.0, 35.1, 34.0, 33.9, 31.9, 31.8, 30.3, 20.2, 19.2. IR (neat) 2958, 1714, 1595 cm−1. LC-MS (M + H)+ calcd for C25H32NO2 378.2433, found 378.2422. Methyl 6-(1-(1,2,3,4-Tetrahydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)vinyl)pyridine-3-carboxylate (8).36 To a suspension of 24 (0.2898 g, 0.797 mmol) in methanol (5.0 mL) was added a solution of KOH (0.116 g) in water (0.18 mL), and the reaction was refluxed at 85 °C for 1 h. The reaction solution was cooled to room temperature and quenched with 20% HCl (26 mL). The crude precipitate was filtered and dried to give a crude white product (0.2753 g, 98%) that was recrystallized from hexanes:ethyl acetate 4:1 to give the pure 8 as a white crystalline solid, mp 243−245 °C. 1H NMR (400 MHz, CDCl3) δ 13.51 (br s, 1H), 9.47 (s, 1H), 8.82 (dd, J = 8.4, 2.0, 1H), 7.41 (d, J = 8.4, 1H), 7.23 (s, 1H), 7.15 (s, 1H), 7.10 (s, 1H), 6.08 (s, 1H), 2.01 (s, 3H), 1.68 (s, 4H), 1.29 (s, 6H), 1.24 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 161.8, 154.3, 146.4, 146.3, 143.6, 139.9, 133.0, 132.5, 130.0, 128.8, 128.6, 128.1, 125.6, 34.8, 34.1, 33.9, 31.8, 31.7, 19.7. IR 2948, 1706, 1590 cm−1. LC-MS (M + H)+ calcd for C23H28NO2 350.2120, found 350.2111. Anal. Calcd for C24H29NO2· HCl: C, 71.58; H, 7.31; N, 3.63. Found: C, 72.64; H, 7.35; N, 3.62. 6-[1-(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydronaphthalen2-yl)cyclopropyl]nicotinic Acid (9).36 To a suspension of 25 (0.3043 g, 0.806 mmol) in methanol (8.0 mL) was added a solution of KOH (0.1287 g) in water (0.18 mL), and the reaction was refluxed at 85 °C for 1 h. The reaction solution was cooled to room temperature and quenched with 20% HCl (27 mL). The crude precipitate was filtered and dried to give a crude white product (0.2543 g, 86%) that was recrystallized from hexanes:ethyl acetate 4:1 to give pure 9 as a white crystalline solid (0.147 g, 50%), mp 273−274 °C. 1H NMR (400 MHz, CDCl3) δ 11.69 (br s, 1H), 9.18 (dd, J = 2.0, 0.8, 1H), 8.05 (dd, J = 8.4, 2.0, 1H), 7.27 (s, 1H), 7.12 (s, 1H), 6.79 (dd, J = 8.4, 0.8, 1H), 2.13 (s, 3H), 1.86−1.87 (m, 2H), 1.69 (s, 4H), 1.39−1.40 (m, 2H), 1.31 (s, 6H), 1.27 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 170.8, 170.2, 151.0, 143.9, 142.7, 137.2, 136.9, 135.7, 129.2, 128.3, 121.4, 120.9, 35.1, 34.0, 33.9, 31.9, 31.8, 30.5, 20.5, 19.2. IR (neat) 2952, 1686, 1593 cm−1. LC-MS (M + H)+ calcd for C24H30NO2 364.2277, found 364.2265. Anal. Calcd for C24H29NO2: C, 79.30; H, 8.04; N, 3.85. Found: C, 78.76; H, 7.93; N, 3.76. Dimethyl Pyrimidine-2,5-dicarboxylate (26). To a 100 mL round-bottom flask charged with pyrimidine-2,5-dicarboxylic acid (5.067 g, 30.1 mmol) was slowly added thionyl chloride (21 mL, 290 mmol) and 3 drops of DMF, and the reaction was refluxed in an oil bath at 85 °C for 3 h. Excess thionyl chloride was removed in vacuo and benzene (20 mL) was added to the crude solid and the benzene was removed in vacuo to give crude pyrimidine-2,5-dicarboyl dichloride that was used without further purification. To the crude pyrimidine-2,5-dicarboyl dichloride was added toluene (13.6 mL), and this solution was added dropwise to a solution of triethylamine (16.5 mL, 118 mmol) in methanol (86 mL). After stirring for 1 h at room temperature, the reaction was quenched with 1 N HCl (120 mL). The reaction was poured into ethyl acetate (80 mL), the layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with saturated NaHCO3 (75 mL) and brine, dried over sodium sulfate, and removed in vacuo to give crude 26. Crude 26 was purified by column chromatography (250 mL SiO2, hexanes:ethyl acetate 2:3) to give 26 (4.26 g, 72%) as a colorless, crystalline solid (mp 140−142 °C). 1H NMR (400 MHz, CDCl3) δ 9.40 (s, 2H), 4.07 (s, 3H), 3.99 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 163.0, 162.9, 158.8, 158.3, 125.4, 53.8, 53.0. IR (neat) 2969, 1709, 1579. 1551 cm−1. GC-MS (M)+ calcd for C8H8N2O4 196.0484, found 196.0485. 8446

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

give a crude brown product that was purified by column chromatography (150 mL SiO2, hexanes:ethyl acetate 97.5:2.5 to 95:5) to give 30 (0.4127 g, 40%) as a white, fiber-like solid, mp 189− 191 °C. 1H NMR (400 MHz, CDCl3) δ 9.25 (s, 2H), 7.17 (s, 1H), 7.11 (s, 1H), 6.83 (d, J = 2.0, 1H), 5.80 (d, J = 2.0, 1H), 3.97 (s, 3H), 1.98 (s, 3H), 1.69 (s, 4H), 1.31 (s, 6H), 1.28 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 168.6, 164.4, 158.2, 148.3, 144.4, 142.1, 136.3, 132.7, 128.0, 127.9, 126.7, 121.1, 52.5, 35.1, 34.0, 33.8, 31.9, 31.8, 20.0. IR (neat) 2959, 1717, 1582 cm−1. LC-MS (M + H)+ calcd for C23H29N2O2 365.2229, found 365.2232. Methyl 2-[(3,5,8,8-Pentamethyl-5.6,7,8-tetrahydronaphthalen-2-yl)cyclopropyl]pyrimidine-5-carboxylate (31). To a suspension of trimethylsulfoxonium iodide (0.365 g, 1.66 mmol) in DMSO (1.2 mL) was added a 20 wt % solution of potassium tertbutoxide in THF (0.94 mL, 1.67 mmol). A solution of 30 (0.4062 g, 1.11 mmol) in THF (4.8 mL) was added dropwise over 20 min at 30− 34 °C with stirring. The reaction was stirred for 60 min at 35 °C, then cooled to room temperature and quenched with 1N HCl (5 mL). The layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated to give an off-white crude solid that was purified by column chromatography (150 mL SiO2, hexanes:ethyl acetate 97.5:2.5 to 90:10) to give 31 (0.2884 g, 68%) as a white crystalline solid, mp 202−203 °C. 1H NMR (400 MHz, CDCl3) δ 9.07 (s, 2H), 7.23 (s, 1H), 7.09 (s, 1H), 3.92 (s, 3H), 2.12 (s, 3H), 1.87−1.88 (m, 2H), 1.67 (s, 4H), 1.46−1.47 (m, 2H), 1.29 (s, 6H), 1.27 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 176.8, 164.7, 157.7, 143.2, 141.8, 136.9, 135.9, 128.6, 127.8, 119.9, 52.3, 35.2, 34.0, 33.9, 31.9, 31.8, 31.7, 21.6, 19.4. IR (neat) 2951, 1719, 1588 cm−1. LCMS (M + H)+ calcd for C24H31N2O2 379.2386, found 379.2385. Methyl 2-(1-(1,2,3,4-Tetrahydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)vinyl)pyridine-5-carboxylate (13). To a suspension of 30 (0.828 g, 2.27 mmol) in methanol (15.0 mL) was added a solution of KOH (0.3627 g) in water (0.54 mL), and the reaction was refluxed at 85 °C for 1 h. The reaction solution was cooled to room temperature and quenched with 20% HCl (60 mL). The crude precipitate was filtered and dried to give a crude white product (0.7741 g, 97%) that was recrystallized from hexanes:ethyl acetate 4:1 to give the pure 13 as a white crystalline solid (0.3869 g, 48%), mp 254−255 °C. 1H NMR (400 MHz, CDCl3) δ 10.11 (br s, 1H), 9.31 (s, 2H), 7.17 (s, 1H), 7.12 (s, 1H), 6.86 (d, J = 1.6, 1H), 5.86 (d, J = 1.6, 1H), 1.99 (s, 3H), 1.68 (s, 4H), 1.28 (s, 6H), 1.27 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 169.0, 168.2, 158.8, 148.0, 144.5, 142.2, 136.0, 132.7, 128.0, 127.9, 127.3, 120.7, 35.1, 34.0, 33.8, 31.9, 31.8, 20.0. IR (neat) 2963, 1682, 1580 cm−1. LC-MS (M + H)+ calcd for C22H27N2O2 351.2073, found 351.2082. Anal. Calcd for C22H26N2O2: C, 75.40; H, 7.48; N, 7.99. Found: C, 75.35; H, 7.54; N, 7.91. 2-[1-(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydronaphthalen2-yl)cyclopropyl]pyrimidine-2-carboxylic Acid (14). To a suspension of 31 (0.287 g, 0.758 mmol) in methanol (5.0 mL) was added a solution of KOH (0.126 g) in water (0.18 mL), and the reaction was refluxed at 85 °C for 1 h. The reaction solution was cooled to room temperature and quenched with 20% HCl (32 mL). The crude precipitate was filtered and dried to give a crude white product (0.2568 g, 92%) that was recrystallized from hexanes:ethyl acetate 4:1 to give the pure 14 as a white crystalline solid (0.2433 g, 88%), mp 266−267 °C. 1H NMR (400 MHz, CDCl3) δ 9.18 (s, 2H), 7.23 (s, 1H), 7.09 (s, 1H), 2.13 (s, 3H), 1.90−1.91 (m, 2H), 1.66 (s, 4H), 1.50−1.51 (m, 2H), 1.27 (s, 6H), 1.26 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 177.6, 168.6, 158.3, 143.3, 141.9, 136.6, 135.9, 128.6, 127.9, 119.2, 35.2, 34.0, 33.9, 32.0, 31.9, 31.8, 22.1, 19.4. IR (neat) 2954, 1679, 1586 cm−1. LC-MS (M + H)+ calcd for C23H29N2O2 365.2229, found 365.2225. Anal. Calcd for C23H28N2O2: C, 75.79; H, 7.74; N, 7.69. Found: C, 75.67; H, 7.95; N, 7.32. 3,4-Dihydro-1,1,4,4,7-pentamethylnaphthalen-2(1H)-one (33).37 A modified procedure of Boehm and co-workers was used.37 To a 100 mL round-bottom flask charged with dihydro-2,2,5,5tetramethylfuran-3(2H)-one (32) (2.4 g, 17 mmol) and toluene (10.0 mL, 94 mmol) at 0 °C was added aluminum chloride (4.55 g, 34 mmol) in one portion with stirring. The reaction was stirred at 0 °C

for 30 min, then warmed to room temperature and stirred for 2 h, and then heated to 50−55 °C with stirring for 15 min. The reaction was cooled to room temperature and then poured onto ice water (50 mL) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo. The resulting crude solid was purified by column chromatography (150 mL SiO2, hexanes:ethyl acetate 98:2) to give 33 (1.6348 g, 44%) as a white crystalline solid, 49−51 °C. 1H NMR (400 MHz, CDCl3) δ 7.26 (d, J = 8.0, 1H), 7.13 (s, 1H), 7.05 (d, J = 8.0, 1H), 2.63 (s, 2H), 2.35 (s, 3H), 1.44 (s, 6H), 1.31 (s, 6H). 13 C NMR (100.6 MHz, CDCl3) δ 214.5, 142.9, 140.6, 136.3, 127.7, 127.4, 124.3, 51.5, 47.9, 37.6, 30.6, 28.4, 21.0. IR (neat) 2968, 1705 cm−1. GC-MS (M)+ calcd for C15H20O 216.1514, found 216.1523. 6-Bromo-3,4-dihydro-1,1,4,4,7-pentamethylnaphthalen2(1H)-one (34). A DCM (22.0 mL) solution of 33 (3.193 g, 14.76 mmol) at 0 °C was stirred during the addition of aluminum chloride (3.993 g, 29.95 mmol), and the mixture was stirred at 0 °C for 5 min. To this solution was added a solution of bromine (0.90 mL, 17.47 mmol) in DCM (11.0 mL) dropwise, and the reaction solution was stirred for 30 min at 0 °C. The reaction solution was poured onto ice (50 mL) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, hexanes:ethyl acetate 98:2) to give 34 (4.36 g, 100%) as a white crystalline solid, mp 114−117 °C. 1H NMR (400 MHz, CDCl3) δ 7.50 (s, 1H), 7.15 (s, 1H), 2.60 (s, 2H), 2.38 (s, 3H), 1.42 (s, 6H), 1.28 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 213.6, 143.2, 142.4, 136.3, 129.5, 128.3, 123.1, 51.2, 47.6, 37.5, 30.4, 28.3, 22.6. IR (neat) 2951, 1706 cm−1. GC-MS (M)+ calcd for C15H19BrO 294.0619, found 294.0617. (E)-1-(6-Bromo-3,4-dihydro-1,1,4,4,7-pentamethylnaphthalen-2(1H)-ylidene)-2-tosylhydrazine (35). To a suspension of 34 (3.0664 g, 10.4 mmol) and p-toluenesulfonylhydrazide (2.2158 g, 11.9 mmol) in methanol (61 mL) was added p-toluenesulfonic acid monohydrate (0.4977 g, 2.616 mmol), and the reaction solution was stirred and refluxed under nitrogen for 24 h. The reaction was then cooled to room temperature and then it was stirred in a salt-water icebath at −15 °C for 1 h and the resulting precipitate was filtered and rinsed with cold methanol to afford 35 as a white crystalline solid (2.41 g, 50%), mp 181−184 °C. 1H NMR (400 MHz, CDCl3) δ 8.03 (br s, 1H), 7.84 (d, J = 8.0, 2H), 7.34 (s, 1H), 7.28 (d, J = 8.4, 2H), 7.12 (s, 1H), 2.41 (s, 2H), 2.38 (s, 3H), 2.33 (s, 3H), 1.37 (s, 6H), 1.08 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 163.2, 144.0, 143.1, 142.7, 136.0, 134.9, 129.3, 128.3, 128.0, 122.7, 42.8, 36.8, 36.2, 29.9, 29.89, 22.5, 21.5. IR (neat) 3251, 2971 cm−1. LC-MS (M + H)+ calcd for C22H28N2O2SBr 463.1055, found 463.1048. 6-Bromo-1,1,4,4,7-pentamethyl-1,4-dihydronaphthalene (36).38 To a suspension of 35 (1.0127 g, 2.19 mmol) in MTBE (20 mL) was added a methyl lithium LiBr complex solution (1.5 M) in ether (4.40 mL, 6.60 mmol) at room temperature with stirring under nitrogen. The solution turned yellow with the evolution of gas (presumably nitrogen), and a fine off-white precipitate formed. The heterogeneous solution was stirred for 1 h, cooled to 0 °C, and then quenched with water (25.0 mL). The reaction was extracted with ethyl acetate, and the organic layers were combined, washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude off-white solid that was purified by column chromatography (150 mL SiO2, hexanes) to give 36 (0.5463 g, 90%) as a white solid (mp 109−111 °C). 1H NMR (400 MHz, CDCl3) δ 7.50 (s, 1H), 7.21 (s, 1H), 5.49 (s, 2H), 2.38 (s, 3H), 1.32 (s, 12H). 13C NMR (100.6 MHz, CDCl3) δ 142.3, 141.8, 135.0, 132.7, 132.6, 129.8, 128.5, 122.4, 35.0, 34.9, 32.5, 32.4, 22.6. IR (neat) 3013, 2958, 1483, 1455, 1079, 889 cm−1. GC-MS (M)+ calcd for C15H19Br 278.0670, found 278.0655. (4-Cyanophenyl)-N-methoxy-N-methylformamide (38).38 The method of Faul and co-workers was used.38 To a suspension of N,O-dimethylhydroxyamine hydrochloride (7.07 g, 72.5 mmol) and K2CO3 (10.0 g, 72.5 mmol) in ACN (100 mL) and water (50 mL) was added 4-cyanobenzoyl chloride (37) (8.00 g, 48.3 mmol), and the reaction was stirred for 2 h at room temperature. The reaction solution 8447

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

was poured into water (50 mL) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated in vacuo to give a crude solid that was purified by column chromatography (250 mL SiO2, hexanes:ethyl acetate 45:55 to 1:1) to give 38 (8.12 g, 88%) as a white crystalline solid, mp 78−79 °C. 1H NMR (400 MHz, CDCl3) δ 7.76 (d, J = 8.4, 2H), 7.69 (d, J = 8.4, 2H), 3.50 (s, 3H), 3.36 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 167.8, 138.2, 131.8, 128.7, 118.1, 114.0, 61.2, 33.0. IR (neat) 2947, 1627 cm−1. GC-MS (M)+ calcd for C10H10N2O2 190.0742, found 190.0739. 4-[(3,5,5,8,8-Pentamethyl-2-5,8-dihydronaphthyl)carbonyl]benzenecarbonitrile (39).38 The method of Faul and co-workers was followed.38 To a solution of 35 (2.00 g, 7.16 mmol) in THF (25 mL) at −78 °C under nitrogen was added a 1.6 M solution of n-BuLi in hexanes (5.40 mL, 8.60 mmol) over 10 min, and the solution was stirred for 20 min at −78 °C. This reaction solution was transferred via airtight syringe to a solution of 38 (1.23 g, 6.47 mmol) in THF (10 mL) at −78 °C, and the combined mixture was stirred for 15 min at −78 °C and then warmed to room temperature before 1.0 N HCl (75 mL) was added to quench the reaction. The solution was poured into ethyl acetate, the layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, hexanes:ethyl acetate 95:5) to give 39 (1.7865 g, 82%) as a white crystalline solid, mp 117−119 °C. 1H NMR (400 MHz, CDCl3) δ 7.91 (d, J = 8.4, 2H), 7.75 (d, J = 8.4, 2H), 5.52 (s, 2H), 2.37 (s, 3H), 1.37 (s, 6H), 1.26 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 196.6, 146.2, 141.6, 139.8, 135.0, 134.4, 132.6, 132.6, 129.2, 128.0, 118.0, 115.9, 35.3, 34.8, 32.4, 32.3, 20.0. IR (neat) 2960, 1669 cm−1. GC-MS (M)+ calcd for C23H23NO 329.1780, found 329.1788. 4-[(3,5,5,8,8-Pentamethyl-2-5,8-dihydronaphthyl)carbonyl]benzoic Acid (40).38 The method of Faul and co-workers was followed.38 To a heterogeneous solution of 39 (1.62 g, 4.92 mmol) in 2-methoxyethanol (20 mL) was added a solution of KOH (1.64 g, 24.5 mmol) in water (10 mL). The reaction was heated in an oil bath at reflux temperature and stirred under nitrogen for 16 h. The reaction was allowed to cool to room temperature before it was quenched with 1 N HCl (50 mL). The solution was poured into ethyl acetate, the layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated in vacuo with additional toluene to azeotrope off 2-methoxyethanol to give 40 (1.65 g, 96%) as a white powder, mp 199−201 °C. 1H NMR (400 MHz, CDCl3) δ 13.36 (br s, 1H), 8.08 (d, J = 8.4, 2H), 7.80 (d, J = 8.4, 2H), 7.42 (s, 1H), 7.34 (s, 1H), 5.54 (s, 2H), 2.25 (s, 3H), 1.33 (s, 6H), 1.22 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 197.0, 166.6, 145.1, 140.7, 139.3, 135.2, 134.5, 133.9, 132.6, 132.5, 129.7, 129.6, 128.8, 127.1, 34.9, 34.5, 32.0, 19.4. IR (neat) 2970, 1745 cm−1. LC-MS (M + H)+ calcd for C23H25O3 349.1804, found 349.1805. 4-[1-(3,5,5,8,8-Pentamethyl-2-5,8-dihydronaphthyl)vinyl]benzoic Acid (10).38 The procedure of Faul and co-workers was followed.38 To a 100 mL round-bottom flask charged with a 3.0 M solution of MeMgCl (1.53 mL, 4.60 mmol) was added THF (3 mL), and the solution was cooled to −10 °C in a salt−water ice bath with stirring under nitrogen. To this solution was added a solution of 40 (0.40 g, 1.15 mmol) in THF (4 mL), dropwise, and the reaction was stirred at 0 °C for 4 h. The reaction was quenched with 1.0 N HCl (15 mL), the solution was extracted with ethyl acetate, and the combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give an intermediate alcohol that was used without further purification. The intermediate alcohol was dissolved in toluene (30 mL), and to this solution was added ptoluenesulfonic acid monohydrate (0.02 g, 0.116 mmol) and the solution was refluxed into a Dean−Stark apparatus prefilled with toluene. After the solution was refluxed for 2 h, it was cooled to room temperature and poured into ethyl acetate and water. The aqueous layer was extracted with ethyl acetate, and the combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by

column chromatography (25 mL SiO2, ethyl acetate) to give 10 (0.3628 g, 91%) as a white crystalline solid (mp 210−213 °C). 1H NMR (400 MHz, CDCl3) δ 11.98 (br s, 1H), 8.05 (d, J = 8.8, 2H), 7.40 (d, J = 8.4, 2H), 7.20 (s, 1H), 7.15 (s, 1H), 5.86 (d, J = 1.2, 1H), 5.54 (s, 2H), 5.38 (d, J = 1.2, 1H), 1.99 (s, 3H), 1.37 (s, 6H), 1.34 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 172.0, 149.0, 146.3, 142.0, 140.1, 138.2, 133.1, 133.1, 133.0, 130.3, 128.0, 127.7, 127.7, 126.6, 117.3, 35.0, 34.9, 32.7, 32.6, 20.0. IR (neat) 2953, 1688, 1606 cm−1. LC-MS (M + H)+ calcd for C24H27O2 347.2011, found 347.1997. Anal. Calcd for C24H26O2: C, 83.20; H, 7.56. Found: C, 82.73; H, 7.54. 6-Bromo-1,2,3,4-tetrahydro-1,1,4,4,7-pentamethylnaphthalene (42).16b The method of Dawson and co-workers was used to synthesize 42.16b To a solution of 22 (1.32 g, 6.52 mmol) in chloroform (6.0 mL) was added bromine (0.5 mL, 9.71 mmol) at room temperature, and the reaction was stirred for 30 min and then diluted with ethyl acetate. The solution was poured into an aqueous saturated solution of Na2SO3, and the biphasic mixture was shaken, the layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated in vacuo to give an oil that was purified by column chromatography (150 mL SiO2, ethyl acetate) to give an inseparable 2:1 mixture of 42:22 (1.679 g, 60% yield for 42) as a white crystalline solid. 1H NMR (400 MHz, CDCl3) δ 7.42 (s, 1H), 7.14 (s, 1H), 2.34 (s, 3H), 1.66 (s, 4H), 1.27 (s, 12H). GC-MS (M)+ calcd for C15H21Br 280.0827, found 280.0812. 5,6,7,8-Tetrahydro-3,5,5,8,8-pentamethylnaphthalen-2-yl-2boronic Acid (43).38 The method of Faul and co-workers was used.38 To a 100 mL round-bottom flask containing THF (30 mL) was added a 1.6 M solution of n-BuLi in hexanes (8.0 mL, 12.8 mmol), and the resulting solution was cooled in a dry ice acetone bath to −78 °C with stirring, under nitrogen. To this solution was added a solution of the 2:1 mixture of 42:22 (3.3587 g, 7.88 mmol) in THF (8 mL) over 20 min and the reaction was stirred at −78 °C for 10 min and a mixture of triisopropylborate (4.9 mL, 21.3 mmol) in THF (10 mL) was added dropwise over 20 min. The reaction was stirred at −78 °C for 1 h and then warmed to room temperature and stirred for 2 h. The reaction was then quenched with 3 N HCl (35 mL), and after stirring for 2 h, it was poured into ethyl acetate, the layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 1:3) to give 43 (0.8838 g, 45%) as a white crystalline solid, mp 220−225 °C. 1 H NMR (400 MHz, CDCl3) δ 8.29 (s, 1H), 7.21 (s, 1H), 2.82 (s, 3H), 1.72 (s, 4H), 1.34 (s, 6H), 1.33 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 149.4, 142.8, 141.4, 136.3, 128.5, 35.1, 35.0, 34.3, 33.8, 31.8, 31.5, 22.6. IR (neat) 2958, 1602, 1335 cm−1. 3-Bromo-4-methylbenzaldehyde (45).43 The method of Adams and co-workers was followed.43 To a solution of 3-bromo-4methylbenzoic acid (44) (5.08 g, 23 mmol) in THF (50 mL) stirring under nitrogen at 0 °C was added a 1 M borane·THF solution (34.6 mL, 34.6 mmol) dropwise. The reaction was warmed to room temperature, stirred for 18 h, then cooled to 0 °C and quenched by the slow addition of water (10 mL). The reaction was warmed to room temperature, and the solvents were removed in vacuo. The crude product was dissolved in ethyl acetate, washed with a 1 M aqueous sodium carbonate solution then brine, and the organic layer was dried over sodium sulfate and concentrated in vacuo to give a benzyl alcohol product (4.75 g, 100%) that was used without further purification. 1H NMR (400 MHz, CDCl3) δ 7.52 (d, J = 0.8, 1H), 7.20 (d, J = 8.0, 1H), 7.17 (dd, J = 7.6, 1.6, 1H), 4.61 (s, 2H), 2.38 (s, 3H), 2.05 (br s, 1H). 13 C NMR (100.6 MHz, CDCl3) δ 140.1, 137.0, 130.8, 130.7, 125.8, 124.9, 64.2, 22.5. To a solution of the benzyl alcohol intermediate (4.5 g, 22.4 mmol) in chloroform (100 mL) was added manganese dioxide (15 g, 172 mmol). The reaction was refluxed with stirring in an oil bath at 70 °C for 18 h. Then it was filtered through Celite, and solvents were removed in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 1:9) to give 45 (3.2974 g, 74%) as a white crystalline solid, mp 45−51 °C. 1H NMR (400 MHz, CDCl3) δ 9.91 (s, 1H), 8.02 (d, J = 8448

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

(E)-Ethyl 3-(3-Bromo-4-(trifluoromethyl)phenyl)acrylate (49). To a solution of a 60% dispersion of NaH in mineral oil (0.31 g, 7.75 mmol) in DME (2 mL) at −30 °C was added a solution of ethyl 2-phosphonoacetate (1.46 mL, 7.29 mmol) in DME (13 mL), and the mixture was stirred at this temperature for 30 min. To this solution was added a solution of 3-bromo-4-(trifluoromethyl)benzaldehyde (48) (1.68 g, 6.63 mmol) in DME (3 mL), and the reaction was stirred at −30 °C for 1.5 h and then poured into water (50 mL) and extracted with ethyl acetate. The combined organic layers were washed with an aqueous saturated NH4Cl solution and then brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 5:95) to give 49 (2.1188 g, 98%) as a colorless crystalline solid, mp 75−76 °C. 1H NMR (400 MHz, CDCl3) δ 7.84 (s, 1H), 7.69 (d, J = 8.0, 1H), 7.59 (d, J = 16.0, 1H), 7.52 (d, J = 8.0, 1H), 6.50, (d, J = 16.0, 1H), 4.27 (q, J = 7.2, 2H), 1.34 (t, J = 7.2, 3H). 13C NMR (100.6 MHz, CDCl3) δ 165.9, 141.0, 139.2, 133.8, 131.4, 130.7, 130.4, 128.3, 128.2, 128.1, 128.1, 126.4, 123.9, 122.2, 121.2, 120.6, 120.5, 60.9, 14.2. IR (neat) 2923, 1711, 1638, 1603 cm−1. GC-MS (M)+ calcd for C12H10F3O2Br 321.9816, found 321.9805. (E)-Ethyl 3-(4-(Trifluoromethyl)-3-(1,2,3,4-tetrahydro1,1,4,4,6-pentamethylnaphthalen-7-yl)phenyl)acrylate (50). To a 50 mL Schlenk flask charged with bromide 49 (1.03 g, 3.20 mmol), boronic acid 43 (0.8040 g, 3.27 mmol), TBAB (1.04 g), Na2CO3 (1.02 g, 9.62 mmol), and water (7.4 mL) was added Pd(OAc)2 (0.0406 g, 0.18 mmol), and the flask was evacuated and backfilled with nitrogen three times. The reaction was stirred at room temperature for 15 min and then placed in an oil bath preheated to 150 °C and stirred for 5 min. The reaction was allowed to cool to room temperature, and the black residue was taken up in ethyl acetate and water. The layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 1:9) to give 50 (1.0138 g, 71%) as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 7.75 (d, J = 8.4, 1H), 7.68 (d, J = 16.0, 1H), 7.57 (d, J = 8.0, 1H), 7.45 (s, 1H), 7.13 (s, 1H), 7.02 (s, 1H), 6.51 (d, J = 16.0, 1H), 4.26 (q, J = 7.2, 2H), 1.98 (s, 3H), 1.69 (s, 4H), 1.32 (t, J = 7.2, 3H), 1.31 (s, 6H), 1.24 (s, 3H), 1.22 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 166.4, 144.5, 142.6, 141.9, 141.3, 137.1, 135.1, 132.3, 131.1, 130.2, 129.9, 127.6, 127.3, 126.7, 126.6, 126.3, 125.0, 122.3, 120.8, 60.7, 35.1, 35.0, 33.9, 33.8, 31.9, 31.8, 31.7, 19.7, 14.2. IR (neat) 2960, 1716, 1641 cm−1. GC-MS (M)+ calcd for C27H31F3O2 444.2276, found 444.2253. (E)-3-(4-(Trifluoromethyl)-3-(1,2,3,4-tetrahydro-1,1,4,4,6pentamethylnaphthalen-7-yl)phenyl)acrylic Acid (16). To a 100 mL round-bottom flask containing 50 (0.4196 g, 1.00 mmol) suspended in methanol (5.0 mL) was added a solution of KOH (0.1706 g, 3.04 mmol) in water (0.22 mL), and the solution was refluxed in an oil bath preheated to 85 °C for 1 h. The reaction was allowed to cool to room temperature and acidified with an aqueous 20% HCl solution (33 mL). The resulting precipitate was filtered and washed with copious amounts of water, and the crude white powder was purified by column chromatography (25 mL SiO2, ethyl acetate:hexanes 15:85) to give 16 (0.3445 g, 88%) as a white crystalline solid, mp 217−221 °C. 1H NMR (400 MHz, CDCl3) δ 7.79 (d, J = 15.6, 1H), 7.78 (d, J = 8.4, 1H), 7.61 (d, J = 8.4, 1H), 7.48 (s, 1H), 7.15 (s, 1H), 7.03 (s, 1H), 6.52, (d, J = 16.0, 1H), 2.00 (s, 3H), 1.70 (s, 4H), 1.36 (s, 6H), 1.25 (s, 3H), 1.23 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 171.7, 145.2, 144.6, 142.1, 141.3, 136.6, 134.9, 132.3, 131.4, 130.7, 130.4, 127.6, 127.3, 126.7, 126.6, 124.9, 122.2, 119.8, 35.1, 35.0, 34.0, 33.8, 31.9, 31.8, 31.7, 19.7. IR (neat) 2961, 1688, 1635 cm−1. LC-MS (M + H)+ calcd for C25H28F3O2 417.2041, found 417.2068. Anal. Calcd for C25H27F3O2: C, 72.10; H, 6.53; F, 13.69. Found: C, 71.42; H, 6.45; F, 14.3. 5,8-Dihydro-3,5,5,8,8-pentamethylnaphthalen-2-yl-2-boronic Acid (51).38 The method of Faul and co-workers was used.38 To a 100 mL round-bottom flask containing THF (20 mL) was added a 1.6 M solution of n-BuLi in hexanes (4.94 mL, 7.90 mmol), and the

1.6, 1H), 7.70 (dd, J = 7.6, 1.6, 1H), 7.39 (d, J = 8.0, 1H), 2.47 (s, 3H). 13 C NMR (100.6 MHz, CDCl3) δ 190.4, 145.1, 135.8, 133.4, 131.3, 128.3, 125.6, 23.4. IR (neat) 2980, 1682, 1598 cm−1. GC-MS (M)+ calcd for C8H7OBr 197.9680, found 197.9665. (E)-Ethyl 3-(3-Bromo-4-methylphenyl)acrylate (46). To a solution of a 60% dispersion of NaH in mineral oil (0.29 g, 7.25 mmol) in DME (2 mL) at −30 °C was added a solution of ethyl 2phosphonoacetate (1.46 mL, 7.29 mmol) in DME (13 mL), and the mixture was stirred at this temperature for 30 min. To this solution was added a solution of 45 (1.32 g, 6.63 mmol) in DME (3 mL), and the reaction was stirred at −30 °C for 1.5 h and then poured into water (50 mL) and extracted with ethyl acetate. The combined organic layers were washed with an aqueous saturated NH4Cl solution and then brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 1:9) to give 46 (1.576 g, 88%) as a colorless crystalline solid, mp 46−47 °C. 1H NMR (400 MHz, CDCl3) δ 7.69 (d, J = 1.6, 1H), 7.57 (d, J = 16.0, 1H), 7.35 (dd, J = 8.0, 1.6, 1H), 7.23 (d, J = 7.6, 1H), 6.38, (d, J = 16.0, 1H), 4.25 (q, J = 7.2, 2H), 2.41 (s, 3H), 1.33 (t, J = 7.2, 3H). 13C NMR (100.6 MHz, CDCl3) δ 166.7, 142.8, 140.1, 133.9, 131.6, 131.1, 126.8, 125.3, 118.6, 60.5, 22.9, 14.2. IR (neat) 2984, 1718, 1698, 1634 cm−1. LC-MS (M + H)+ calcd for C12H14O2Br 269.0177, found 269.0171. (E)-Ethyl 3-(3-(1,2,3,4-Tetrahydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)-4-methylphenyl)acrylate (47). To a 50 mL Schlenk flask charged with bromide 46 (0.4317 g, 1.60 mmol), boronic acid 43 (0.4010 g, 1.63 mmol), TBAB (0.52 g), Na2CO3 (0.51 g, 4.81 mmol), and water (3.7 mL), was added Pd(OAc)2 (0.0203 g, 0.09 mmol), and the flask was evacuated and backfilled with nitrogen three times. The reaction was stirred at room temperature for 15 min and then placed in an oil bath preheated to 150 °C and stirred for 5 min. The reaction was allowed to cool to room temperature, and the black residue was taken up in ethyl acetate and water. The layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 1:9) to give 47 (0.5032 g, 80%) as a colorless oil. 1 H NMR (400 MHz, CDCl3) δ 7.68 (d, J = 16.0, 1H), 7.41 (dd, J = 8.0, 2.0, 1H), 7.33 (d, J = 1.6, 1H), 7.27 (d, J = 8.0, 1H), 7.16 (s, 1H), 7.00 (s, 1H), 6.39, (d, J = 16.0, 1H), 4.25 (q, J = 7.2, 2H), 2.09 (s, 3H), 2.01 (s, 3H), 1.70 (s, 4H), 1.33 (t, J = 7.2, 3H), 1.32 (s, 6H), 1.26 (s, 3H), 1.24 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 171.1, 167.2, 144.6, 143.8, 143.1, 142.5, 142.1, 141.6, 139.0, 138.8, 137.7, 132.8, 132.3, 131.7, 130.3, 129.3, 127.9, 127.6, 127.4, 127.2, 126.6, 117.2, 60.4, 60.3, 35.2, 35.1, 34.0, 33.9, 32.0, 31.9, 31.8, 31.8, 21.0, 20.0, 19.8, 19.5, 14.3, 14.1. IR (neat) 2957, 1711, 1635 cm−1. LC-MS (M + H)+ calcd for C27H35O2 391.2637, found 391.2655. (E)-3-(3-(1,2,3,4-Tetrahydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)-4-methylphenyl)acrylic Acid (15). To a 100 mL roundbottom flask containing 47 (0.3288 g, 0.84 mmol) suspended in methanol (5.0 mL) was added a solution of KOH (0.1412 g, 2.5 mmol) in water (0.18 mL), and the solution was refluxed in an oil bath preheated to 85 °C for 1 h. The reaction was allowed to cool to room temperature and acidified with an aqueous 20% HCl solution (28 mL). The resulting precipitate was filtered and washed with copious amounts of water, and the crude white powder was purified by column chromatography (25 mL SiO2, ethyl acetate:hexanes 15:85) to give 15 (0.2295 g, 75%) as a white crystalline solid, mp 189−196 °C. 1H NMR (400 MHz, CDCl3) δ 7.79 (d, J = 16.0, 1H), 7.44 (dd, J = 8.0, 1.6, 1H), 7.37 (d, J = 1.6, 1H), 7.29 (d, J = 7.6, 1H), 7.17 (s, 1H), 7.01 (s, 1H), 6.42, (d, J = 16.0, 1H), 2.11 (s, 3H), 2.02 (s, 3H), 1.71 (s, 4H), 1.33 (s, 3H), 1.32 (s, 3H), 1.27 (s, 3H), 1.24 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 172.6, 147.1, 143.8, 142.7, 142.1, 139.7, 137.5, 132.3, 131.4, 130.4, 129.6, 127.7, 126.9, 116.3, 35.2, 35.1, 34.0, 33.9, 32.0, 31.9, 31.8, 20.1, 19.5. IR (neat) 2957, 1680, 1627 cm−1. LC-MS (M + H)+ calcd for C25H31O2 363.2324, found 363.2311. Anal. Calcd for C25H30O2: C, 82.83; H, 8.34. Found: C, 81.84; H, 8.28. 8449

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

resulting solution was cooled in a dry ice acetone bath to −78 °C with stirring, under nitrogen. To this solution was added a solution of 36 (2.1244 g, 7.61 mmol) in THF (5 mL) over 20 min and the reaction was stirred at −78 °C for 10 min and a mixture of triisopropylborate (3.5 mL, 15.2 mmol) in THF (5 mL) was added dropwise over 20 min. The reaction was stirred at −78 °C for 2 h and then warmed to room temperature and stirred for 1 h. The reaction was then quenched with 3 N HCl (30 mL), and after stirring for 30 min, it was poured into ethyl acetate, the layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 1:3) to give 51 (1.1639 g, 62%) as a white crystalline solid, mp 162−166 °C. 1H NMR (400 MHz, CDCl3) δ 8.36 (s, 1H), 7.28 (s, 1H), 5.56 (s, 2H), 2.87 (s, 3H), 1.41 (s, 6H), 1.39 (s, 6H). IR (neat) 3213 (br), 2958, 1605 cm−1. (2E)-Ethyl 3-(3-(1,4-Dihydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)-4-methylphenyl)acrylate (52). To a 50 mL Schlenk flask charged with bromide 46 (0.6506 g, 2.41 mmol), boronic acid 51 (0.2032 g, 0.83 mmol), TBAB (0.26 g), Na2CO3 (0.256 g, 2.42 mmol), and water (1.85 mL) was added Pd(OAc)2 (0.0136 g, 0.061 mmol), and the flask was evacuated and backfilled with nitrogen three times. The reaction was stirred at room temperature for 15 min and then placed in an oil bath preheated to 150 °C and stirred for 5 min. The reaction was allowed to cool to room temperature, and the black residue was taken up in ethyl acetate and water. The layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 2.5:97.5) to give 52 (0.2256 g, 72%) as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 7.69 (d, J = 16.0, 1H), 7.42 (dd, J = 8.0, 2.0, 1H), 7.36 (d, J = 2.0, 1H), 7.29 (d, J = 8.0, 1H), 7.22 (s, 1H), 7.06 (s, 1H), 6.41, (d, J = 16.0, 1H), 5.53 (s, 2H), 4.25 (q, J = 7.2, 2H), 2.10 (s, 3H), 2.04 (s, 3H), 1.38 (s, 6H), 1.33 (t, J = 7.2, 3H), 1.32 (s, 3H), 1.30 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 167.8, 144.5, 142.4, 141.5, 139.8, 139.0, 138.1, 133.1, 133.0, 132.7, 131.8, 130.4, 129.3, 127.3, 126.8, 126.6, 117.3, 60.3, 35.0, 34.9, 32.7, 32.6, 32.6, 20.0, 19.6, 14.3, 14.1. IR (neat) 2948, 1698, 1634 cm−1. GC-MS (M)+ calcd for C27H32O2 388.2402, found 388.2414. (2E)-3-(3-(1,4-Dihydro-1,1,4,4,6-pentamethylnaphthalen-7yl)-4-methylphenyl)acrylic Acid (17). To a 100 mL round-bottom flask containing 52 (0.5555 g, 1.43 mmol) suspended in methanol (5.0 mL) was added a solution of KOH (0.2454 g, 4.37 mmol) in water (0.30 mL), and the solution was refluxed in an oil bath preheated to 85 °C for 1 h. The reaction was allowed to cool to room temperature and acidified with an aqueous 20% HCl solution (50 mL). The resulting precipitate was filtered and washed with copious amounts of water, and the crude white powder was purified by column chromatography (25 mL SiO2, ethyl acetate:hexanes 15:85 to 2:3) to give 17 (0.3917 g, 76%) as a white crystalline solid, mp 183−185 °C. 1H NMR (400 MHz, CDCl3) δ 7.80 (d, J = 15.6, 1H), 7.46 (dd, J = 8.0, 2.0, 1H), 7.39 (d, J = 1.6, 1H), 7.31 (d, J = 8.0, 1H), 7.24 (s, 1H), 7.07 (s, 1H), 6.43, (d, J = 16.0, 1H), 5.54 (s, 2H), 2.12 (s, 3H), 2.05 (s, 3H), 1.39 (s, 6H), 1.33 (s, 3H), 1.31 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 172.6, 147.8, 142.5, 141.6, 139.9, 137.9, 133.1, 133.0, 132.7, 131.4, 130.5, 129.6, 127.4, 127.0, 126.8, 116.3, 35.0, 34.9, 32.7, 32.6, 32.6, 20.1, 19.6. IR (neat) 2956, 1689, 1624 cm−1. GC-MS (M)+ calcd for C25H28O2 360.2089, found 360.2089. Anal. Calcd for C25H27O2: C, 83.29; H, 7.83. Found: C, 82.75; H, 7.83. 2-Bromo-4-formylphenyl Acetate (54).39 The method of Gronemeyer and co-workers was followed.39 To a 100 mL roundbottom flask charged with 3-bromo-4-hydroxybenzaldehyde (2.018 g, 10.0 mmol) was added DMAP (0.066 g, 0.54 mmol) and acetic anhydride (11.0 mL, 116 mmol), a reflux condenser was appended, the apparatus was evacuated and backfilled with nitrogen, and the solution was heated to 135 °C in a preheated oil bath for 8 min. The reaction was cooled to room temperature and then poured into water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in

vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 1:9) to give 54 (2.44 g, 90%) as a white crystalline solid, mp 58−60 °C. 1H NMR (400 MHz, CDCl3) δ 9.94 (s, 1H), 8.13 (d, J = 2.0, 1H), 7.85 (dd, J = 8.4, 2.0, 1H), 7.31 (d, J = 8.4, 1H), 2.38 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 189.5, 167.8, 152.8, 135.2, 134.5, 129.8, 124.5, 117.4, 20.7. IR (neat) 2944, 1743, 1697, 1686 cm−1. GC-MS (M)+ calcd for C9H7O3Br 241.9579, found 241.9574. (E)-Ethyl 3-(4-Acetoxy-3-bromophenyl)acrylate (55).39 The method of Gronemeyer and co-workers was followed.39 To a solution of a 60% dispersion of NaH in mineral oil (0.31 g, 7.75 mmol) in DME (2 mL) at −30 °C was added a solution of ethyl 2phosphonoacetate (1.46 mL, 7.29 mmol) in DME (13 mL), and the mixture was stirred at this temperature for 30 min. To this solution was added a solution of 54 (1.61 g, 6.62 mmol) in DME (3 mL), and the reaction was stirred at −30 °C for 1.5 h and then poured into water (50 mL) and extracted with ethyl acetate. The combined organic layers were washed with an aqueous saturated NH4Cl solution and then brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 1:9) to give 55 (1.7598 g, 84%) as a colorless crystalline solid, mp 71−72 °C. 1H NMR (400 MHz, CDCl3) δ 7.76 (d, J = 2.0, 1H), 7.58 (d, J = 16.0, 1H), 7.46 (dd, J = 8.0, 2.0, 1H), 7.14 (d, J = 8.4, 1H), 6.38, (d, J = 16.0, 1H), 4.25 (q, J = 7.2, 2H), 2.36 (s, 3H), 1.33 (t, J = 7.2, 3H). 13C NMR (100.6 MHz, CDCl3) δ 168.2, 166.4, 149.3, 141.9, 133.9, 132.5, 127.9, 124.0, 119.8, 116.8, 60.6, 20.7, 14.2. IR (neat) 2981, 1758, 1715 cm−1. GC-MS (M)+ calcd for C13H13O2Br 311.9997, found 311.9988. (2E)-Ethyl-3-(4-acetoxy-3-(1,4-dihydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)phenyl)acrylate (56). To a 50 mL Schlenk flask charged with bromide 55 (0.5038 g, 1.60 mmol), boronic acid 51 (0.4040 g, 1.65 mmol), TBAB (0.52 g), Na2CO3 (0.51 g, 4.81 mmol), and water (3.70 mL) was added Pd(OAc)2 (0.0277 g, 0.123 mmol), and the flask was evacuated and backfilled with nitrogen three times. The reaction was stirred at room temperature for 15 min and then placed in an oil bath preheated to 150 °C and stirred for 5 min. The reaction was allowed to cool to room temperature, and the black residue was taken up in ethyl acetate and water. The layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 5:95 to 3:7) to give 56 (0.3673 g, 54%) as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 7.69 (d, J = 16.0, 1H), 7.54 (dd, J = 8.0, 2.0, 1H), 7.50 (d, J = 2.0, 1H), 7.23 (s, 1H), 7.17 (d, J = 8.4, 1H), 7.10 (s, 1H), 6.41, (d, J = 16.0, 1H), 5.52 (s, 2H), 4.25 (q, J = 7.2, 2H), 2.13 (s, 3H), 1.91 (s, 3H), 1.36 (s, 6H), 1.33 (t, J = 7.2, 3H), 1.30 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 169.1, 166.8, 149.9, 143.5, 142.1, 139.6, 135.6, 133.5, 133.3, 133.0, 132.9, 132.2, 131.0, 127.8, 127.6, 127.5, 123.1, 118.5, 60.5, 35.0, 34.9, 32.6, 20.4, 19.6, 14.2. IR (neat) 3317, 2970, 1680, 1632 cm−1. LC-MS (M + H)+ calcd for C28H33O4 433.2379, found 433.2371. (2E)-3-(3-(1,4-Dihydro-1,1,4,4,6-pentamethylnaphthalen-7yl)-4-hydroxyphenyl)acrylic Acid (18). To a 100 mL roundbottom flask containing 56 (0.3622 g, 0.87 mmol) suspended in methanol (5.0 mL) was added a solution of KOH (0.3326 g, 5.93 mmol) in water (0.48 mL), and the solution was refluxed in an oil bath preheated to 85 °C for 1 h. The reaction was allowed to cool to room temperature and acidified with an aqueous 20% HCl solution (50 mL). The resulting precipitate was filtered and washed with copious amounts of water, and the crude white powder was purified by column chromatography (25 mL SiO2, ethyl acetate:hexanes 10:90 to 2:5) to give 18 (0.2820 g, 89%) as a white crystalline solid, mp 163−168 °C. 1 H NMR (400 MHz, CDCl3) δ 7.76 (d, J = 15.6, 1H), 7.51 (dd, J = 8.4, 2.0, 1H), 7.39 (d, J = 2.4, 1H), 7.39 (s, 1H), 7.21 (s, 1H), 7.03 (d, J = 8.4, 1H), 6.34, (d, J = 16.0, 1H), 5.54 (s, 2H), 2.15 (s, 3H), 1.39 (s, 6H), 1.33 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 172.6, 155.2, 146.7, 143.4, 141.2, 134.2, 132.9, 132.8, 132.0, 130.8, 129.5, 128.6, 128.5, 128.0, 126.6, 115.9, 114.7, 60.4, 35.1, 35.0, 32.6, 32.5, 21.0, 19.4, 14.1. IR (neat) 2959, 1682, 1629 cm−1. LC-MS (M + H)+ calcd for 8450

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

then brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 5:95) to give 61 (1.6796 g, 98%) as a colorless crystalline solid, mp 34−36 °C. 1H NMR (400 MHz, CDCl3) δ 7.66 (t, J = 1.6 Hz, 1H), 7.59 (d, J = 16.0, 1H), 7.49 (dq, J = 7.6, 0.8 Hz, 1H), 7.42 (d, J = 7.6, 0.8 Hz, 1H), 7.25 (d, J = 8.0 Hz, 1H), 6.42 (d, J = 16.0, 1H), 4.27 (q, J = 7.2, 2H), 1.33 (t, J = 7.2, 3H). 13C NMR (100.6 MHz, CDCl3) δ 166.5, 142.8, 136.5, 132.9, 130.6, 130.3, 126.6, 122.9, 119.7, 60.6, 14.2. IR (neat) 2978, 1708, 1640 cm−1. GC-MS (M)+ calcd for C11H11BrO2 253.9942, found 253.9946. (E)-Ethyl 3-(3-(1,2,3,4-Tetrahydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)phenyl)acrylate (62). To a 50 mL Schlenk flask charged with bromide 61 (0.4125 g, 1.61 mmol), boronic acid 43 (0.4020 g, 1.64 mmol), TBAB (0.52 g), Na2CO3 (0.52 g, 4.81 mmol), and water (3.7 mL), was added Pd(OAc)2 (0.0203 g, 0.09 mmol), and the flask was evacuated and backfilled with nitrogen three times. The reaction was stirred at room temperature for 15 min and then placed in an oil bath preheated to 150 °C and stirred for 5 min. The reaction was allowed to cool to room temperature, and the black residue was taken up in ethyl acetate and water. The layers were separated, and the aqueous layer was extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude product that was purified by column chromatography (150 mL SiO2, ethyl acetate:hexanes 1:9) to give 62 (0.4295 g, 70%) as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 7.73 (d, J = 16.0, 1H), 7.51−7.49 (m, 2H), 7.45−7.36 (m, 2H), 7.21 (s, 1H), 7.16 (s, 1H), 6.46 (d, J = 16.0, 1H), 4.27 (q, J = 7.2, 2H), 2.24 (s, 3H), 1.71 (s, 4H), 1.36−1.26 (m, 15H). 13C NMR (100.6 MHz, CDCl3) δ 167.0, 144.6, 144.2, 142.8, 142.5, 138.3, 134.2, 132.1, 131.2, 129.0, 128.5, 128.3, 127.8, 126.1, 118.3, 60.5, 35.1, 33.9, 33.9, 31.9, 31.8, 21.0, 20.1, 14.3, 14.1. IR (neat) 2957, 1711, 1637 cm−1. GC-MS (M)+ calcd for C26H32O2 376.2402, found 376.2390. (E)-3-(3-(1,2,3,4-Tetrahydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)phenyl)acrylic Acid (12). To a 100 mL round-bottom flask containing 62 (0.400 g, 1.06 mmol) suspended in methanol (5.0 mL) was added a solution of KOH (0.2002 g, 3.57 mmol) in water (0.26 mL), and the solution was refluxed in an oil bath preheated to 85 °C for 1 h. The reaction was allowed to cool to room temperature and acidified with an aqueous 20% HCl solution (30 mL). The resulting precipitate was filtered and washed with copious amounts of water, and the crude white powder was purified by column chromatography (25 mL SiO2, ethyl acetate:hexanes 15:85) to give 12 (0.3119 g, 84%) as a white crystalline solid, 205−207 °C. 1H NMR (400 MHz, CDCl3) δ 7.85 (d, J = 16.0 Hz, 1H), 7.54−7.53 (m, 2H), 7.46 (t, J = 7.6, 1H), 7.41 (dt, J = 8.4, 1.6 Hz, 1H), 7.22 (s, 1H), 7.17 (s, 1H), 6.49, (d, J = 16.0, 1H), 2.25 (s, 3H), 1.73 (s, 4H), 1.33 (s, 6H), 1.31 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 172.2, 147.2, 144.3, 143.0, 142.6, 138.1, 133.8, 132.1, 131.8, 129.3, 128.6, 128.4, 127.8, 126.5, 117.3, 35.1, 34.0, 33.9, 31.9, 31.8, 20.1. IR (neat) 2955, 1706, 1630 cm−1. LC-MS (M + H)+ calcd for C24H29O2 349.2168, found 349.2168. Anal. Calcd for C24H28O2: C, 82.72; H, 8.10. Found: C, 82.05; H, 7.94.

C24H27O3 363.1960, found 363.1967. Anal. Calcd for C24H26O3: C, 79.53; H, 7.23. Found: C, 78.33; H, 7.14. 1-(4-(Methoxycarbonyl)-3-fluorophenyl)vinyl Trifluoromethanesulfonate (58). Following a procedure similar to that used by Faul and co-workers,38 to a solution of methyl 4-acetyl-2fluorobenzoate (57) (1.972 g, 10.05 mmol) in dichloromethane (15.0 mL) was added finely ground anhydrous Na2CO3 (1.71 g, 16.1 mmol) followed by trifluoromethanesulfonic anhydride (3.4 mL, 20 mmol). The reaction was stirred under nitrogen for 24 h, at which point an additional amount of finely ground anhydrous Na2CO3 (0.42 g, 4.0 mmol) followed by trifluoromethanesulfonic anhydride (1.7 mL, 10 mmol) was added, and the reaction was stirred for an additional 48 h. The mixture was filtered, concentrated in vacuo to an oil, and loaded directly onto a silica gel column (2.5% ethyl acetate in hexanes). The product-containing fractions were combined to give 58 as a crystalline solid (0.9011 g, 27%). 1H NMR (400 MHz, CDCl3) δ 8.00 (t, J = 8.0, 1H), 7.38 (dd, J = 8.0, 1.6, 1H), 7.31 (dd, J = 11.2, 1.6, 1H), 5.76 (d, J = 4.0, 1H), 5.55 (d, J = 4.4, 1H), 3.94 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 163.9, 163.9, 163.1, 160.5, 150.9, 150.9, 137.9, 137.8, 132.9, 120.6, 120.5, 120.0, 119.9, 116.8, 114.0, 113.8, 107.4, 52.6. IR (neat) 2970, 1691, 1414 cm−1. LC-MS (M)+ calcd for C11H9F4O5S 329.0107, found 329.0106. Methyl 2-Fluoro-4-(1-(1,4-dihydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)vinyl)benzoate (59). A solution of boronic acid (51) (0.294 g, 1.20 mmol), triflate 58 (0.429 g, 1.31 mmol), Pd(OAc)2 (0.014 g, 0.062 mmol), P(o-Tol)3 (0.030 g, 0.099 mmol), and Et3N (0.34 mL, 2.4 mmol) in DMF (4.0 mL) was heated to 50 °C and stirred for 2 h. After cooling to room temperature, the reaction was poured into water and extracted with ethyl acetate. The combined organic layers were washed with water and brine and then dried over sodium sulfate, filtered, and concentrated in vacuo to give a crude oil that was purified by column chromatography (silica gel, 2.5% ethyl acetate in hexanes) to give 59 as a white, crystalline solid: (0.252 g, 55%), mp 124−126 °C. 1H NMR (400 MHz, CDCl3) δ 7.88 (t, J = 8.0, 1H). 7.17−7.14 (m, 3H), 7.03 (dd, J = 12.4, 1.6, 1H), 5.85 (d, J = 1.2, 1H), 5.53 (s, 2H), 5.39 (d, J = 1.2, 1H), 3.92 (s, 3H), 1.98 (s, 3H), 1.37 (s, 6H), 1.34 (s, 3H). 13C NMR (100.6 MHz, CDCl3) δ 164.7, 164.7, 163.3, 160.7, 148.0, 147.9, 147.7, 147.6, 142.3, 140.2, 137.6, 133.0, 133.0, 1132.0, 127.8, 127.6, 122.0, 122.0, 117.8, 117.1, 117.0, 114.9, 114.7, 52.2, 35.0, 34.9, 32.6, 32.6, 19.9. IR (neat) 2952, 1712, 1618 cm−1. LC-MS (M + H)+ calcd for C25H28FO2 379.2073, found 379.2069. 2-Fluoro-4-(1-(1,4-dihydro-1,1,4,4,6-pentamethylnaphthalen-7-yl)vinyl)benzoic Acid (19). To a suspension of 59 (0.2475 g, 0.65 mmol) in methanol (3.4 mL) was added a solution of KOH (0.1009 g) in water (0.18 mL), and the reaction was refluxed at 85 °C for 1 h. The reaction solution was cooled to room temperature and quenched with 1N HCl (50 mL). The crude precipitate was filtered and dried to give a crude white product (0.2135 g, 89%) that was purified by column chromatography (silica gel, 10−30% ethyl acetate in hexanes) to give 19 as a crystalline solid (0.1664 g, 70%), mp 187− 188 °C. 1H NMR (400 MHz, CDCl3) δ 11.29 (br s, 1H), 7.97 (t, J = 8.0, 1H), 7.20−7.16 (m, 3H), 7.07 (dd, J = 12.4, 1.2, 1H), 5.88 (d, J = 0.8, 1H), 5.54 (s, 2H), 5.42 (d, J = 0.8, 1H), 2.00 (s, 3H), 1.37 (s, 6H), 1.34 (s, 6H). 13C NMR (100.6 MHz, CDCl3) δ 169.5, 169.4, 164.0, 161.4, 148.8, 148.7, 147.9, 142.4, 140.2, 137.5, 133.0, 133.0, 132.7, 127.8, 127.6, 122.1, 122.1, 118.3, 116.0, 115.9, 115.1, 114.8, 35.0, 34.9, 32.6, 32.6, 19.9. IR (neat) 2958, 1701, 1617 cm−1. LC-MS (M + H)+ calcd for C24H26O2F 365.1917, found 365.1923. Anal. Calcd for C24H25O2F: C, 79.09; H, 6.91; F, 5.21. Found: C, 78.74; H, 6.76; F, 5.10. (E)-Ethyl 3-(3-Bromophenyl)acrylate (61). To a solution of a 60% dispersion of NaH in mineral oil (0.31 g, 7.75 mmol) in DME (2 mL) at −30 °C was added a solution of ethyl 2-phosphonoacetate (1.46 mL, 7.29 mmol) in DME (13 mL), and the mixture was stirred at this temperature for 30 min. To this solution was added a solution of 3-bromobenzaldehyde (60) (1.23 g, 6.65 mmol) in DME (3 mL), and the reaction was stirred at −30 °C for 1.5 h and then poured into water (50 mL) and extracted with ethyl acetate. The combined organic layers were washed with an aqueous saturated NH4Cl solution and



ASSOCIATED CONTENT

S Supporting Information *

1

H NMR and 13C NMR spectra of all compounds reported in the experimentals and dose−response curves for all novel and reported rexinoids. The X-ray data for compound 14 (CCDC 942622) can be obtained free of charge from The Cambridge Crystallographic Data Centre via www.ccdc.cam.ac.uk/data_ request/cif. This material is available free of charge via the Internet at http://pubs.acs.org.



AUTHOR INFORMATION

Corresponding Author

*Phone: 602-543-6937. Fax: 602-543-6073. E-mail: Carl. [email protected]. 8451

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Author Contributions ∥

(10) (a) Lemon, B. D.; Freedman, L. P. Selective effects of ligands on vitamin D3 receptor- and retinoid X receptor-mediated gene activation in vivo. Mol. Cell. Biol. 1996, 16, 1006−1016. (b) MacDonald, P. N.; Dowd, D. R.; Nakajima, S.; Galligan, M. A.; Reeder, M. C.; Haussler, C. A.; Ozato, K.; Haussler, M. R. Retinoid X receptors stimulate and 9cis retinoic acid inhibits 1,25-dihydroxyvitamin D3-activated expression of the rat osteocalcin gene. Mol. Cell. Biol. 1993, 13, 5907−5917. (c) Thompson, P. D.; Jurutka, P. W.; Haussler, C. A.; Whitfield, G. K.; Haussler, M. R. Heterodimeric DNA binding by the vitamin D receptor and retinoid X receptors is enhanced by 1,25-dihydroxyvitamin D3 and inhibited by 9-cis retinoic acid: evidence for allosteric receptor interactions. J. Biol. Chem. 1998, 273, 8483−8491. (11) Lehmann, J. M.; Zhang, X. K.; Graupner, G.; Lee, M. O.; Hermann, T.; Hoffmann, B.; Pfahl, M. Formation of Retinoid X Receptor Homodimers Leads to Repression of T3 Response: Hormonal Cross Talk by Ligand-Induced Squelching. Mol. Cell. Biol. 1993, 13, 7698−7707. (12) (a) Raval-Pandya, M.; Freedman, L. P.; Li, H.; Christakos, S. Thyroid Hormone Receptor Does Not Heterodimerize with the Vitamin D Receptor but Represses Vitamin D Receptor-Mediated Transactivation. Mol. Endocrinol. 1998, 12, 1367−1379. (b) Thompson, P. D.; Hsieh, J.-C.; Whitfield, G. K.; Haussler, C. A.; Jurutka, P. W.; Galligan, M. A.; Tillman, J. B.; Spindler, S. R.; Haussler, M. R. Vitamin D receptor displays DNA binding and transactivation as a heterodimer with the retinoid X receptor, but not with the thyroid hormone receptor. J. Cell. Biochem. 1999, 75, 462−480. (13) Yen, P. M.; Liu, Y.; Sugawara, A.; Chin, W. W. Vitamin D Receptors Repress Basal Transcription and Exert Dominant Negative Activity on Triiodothyronine-Mediated Transcriptional Activity. J. Biol. Chem. 1996, 271, 10910−10916. (14) Altucci, L.; Leibowitz, M. D.; Ogilvie, K. M.; de Lera, A. R.; Gronemeyer, H. RAR and RXR modulation in cancer and metabolic disease. Nature Rev. Drug Discovery 2007, 6, 793−810. (15) Lehmann, J. M.; Jong, L.; Fanjul, A.; Cameron, J. F.; Lu, X.-P.; Haefner, P.; Dawson, M. I.; Pfahl, M. Retinoids selective for retinoid X receptor pathways. Science 1992, 258, 1944−1946. (16) (a) Jong, L.; Lehmann, J. M.; Hobbs, P. D.; Harlev, E.; Huffman, J. C.; Pfahl, M.; Dawson, M. I. Conformational effects on retinoid receptor selectivity. 1. Effect of 9-double bond geometry on retinoid X receptor activity. J. Med. Chem. 1993, 36, 2605−2613. (b) Dawson, M. I.; Jong, L.; Hobbs, P. D.; Cameron, J. F.; Chao, W.-R.; Pfahl, M.; Lee, M.-O.; Shroot, B.; Pfahl, M. Conformational Effects on Retinoid Receptor Selectivity. 2. Effects of Retinoid Bridging Group on Retinoid X Receptor Activity and Selectivity. J. Med. Chem. 1995, 38, 3368−3383. (17) Boehm, M. F.; Zhang, L.; Badea, B. A.; White, S. K.; Mais, D. E.; Berger, E.; Suto, C. M.; Goldman, M. E.; Heyman, R. A. Synthesis and Structure−Activity Relationships of Novel Retinoid X ReceptorSelective Retinoids. J. Med. Chem. 1994, 37, 2930−2941. (18) Daiss, J. O.; Burschka, C.; Mills, J. S.; Montana, J. G.; Showell, G. A.; Fleming, I.; Gaudon, C.; Ivanova, D.; Gronemeyer, H.; Tacke, R. Synthesis, Crystal Structure Analysis, and Pharmacological Characterization of Disila-bexarotene, a Disila-Analogue of the RXRSelective Retinoid Agonist Bexarotene. Organometallics 2005, 24, 3192−3199. (19) Cesario, R. M.; Stone, J.; Yen, W.-C.; Bissonnette, R. P.; Lamph, W. W. Differentiation and growth inhibition mediated via the RXR:PPARγ heterodimer in colon cancer. Cancer Lett. 2006, 240, 225−233. (20) Yen, W. -C.; Prudente, R. Y.; Lamph, W. W. Synergistic effect of a retinoid X receptor-selective ligand bexarotene (LGD1069, Targretin) and paclitaxel (Taxol) in mammary carcinoma. Breast Cancer Res. Treat. 2004, 88, 141−148. (21) (a) Yen, W.-C.; Corpuz, M. R.; Prudente, R. Y.; Cooke, T. A.; Bissonnette, R. P.; Negro-Vilar, A.; Lamph, W. W. A Selective Retinoid X Receptor Agonist Bexarotene (Targretin) Prevents and Overcomes Acquired Paclitaxel (Taxol) Resistance in Human Non-Small Cell Lung Cancer. Clin. Cancer Res. 2004, 10, 8656−8664. (b) Dragnev, K. H.; Petty, W. J.; Shah, S. J.; Lewis, L. D.; Black, C. C.; Memoli, V.;

These authors contributed equally to this work.

Notes

The authors declare the following competing financial interest(s): Patent applications covering the technologies described in this work have been applied for on behalf of the Arizona Board of Regents.



ACKNOWLEDGMENTS We thank the Thome Foundation and the U.S. National Institutes of Health for financial support of this work (1 R15 CA139364-01A2). Thanks are also given to Dr. Natalya Zolotova of the High-Resolution Mass Spectrometry Laboratory at Arizona State University (ASU), Tempe (USA). Computer time was provided by USF Research Computing and XSEDE. Patent applications covering the technologies described in this work have been applied for on behalf of the Arizona Board of Regents.



ABBREVIATIONS USED RXR, retinoid X receptor; RAR, retinoic acid receptor; CTCL, cutaneous T-cell lymphoma; RXRE, retinoid X receptor element; HRE, hormone responsive element; TR, thyroid hormone receptor; VDR, vitamin D receptor; SNuRMs, specific nuclear receptor modulators; NR, nuclear receptor; LXR, liver X receptor; PPAR, peroxisome proliferator activating receptor; LBD, ligand binding domain; NaBu, sodium butyrate



REFERENCES

(1) (a) Mangelsdorf, D. J.; Umesono, K.; Evans, R. M. The Retinoid Receptors. In The Retinoids; Academic Press: Orlando, FL, 1994; pp 319−349. (b) Leid, M.; Kastner, P.; Chambon, P. Multiplicity Generates Diversity in the Retinoic Acid Signaling Pathways. Trends Biochem. Sci. 1992, 17, 427−433. (2) Olefsky, J. M. Nuclear Receptor Minireview Series. J. Biol. Chem. 2001, 276, 36863−36864. (3) Forman, B. M.; Yang, C.-R.; Au, M.; Casanova, J.; Ghysdael, J.; Samuels, H. H. A domain containing leucine-zipper-like motifs mediates novel in vitro interactions between the thyroid hormone and retinoic acid receptors. Mol. Endocrinol. 1989, 3, 1610−1626. (4) Mangelsdorf, D. J.; Evans, R. M. The RXR heterodimers and orphan receptors. Cell 1995, 83, 841−850. (5) Thompson, P. D.; Remus, L. S.; Hsieh, J.-C.; Jurutka, P. W.; Whitfield, G. K.; Galligan, M. A.; Encinas Dominguez, C.; Haussler, C. A.; Haussler, M. R. Distinct retinoid X receptor activation function-2 residues mediate transactivation in homodimeric and vitamin D receptor heterodimeric contexts. J. Mol. Endocrinol. 2001, 27, 211− 227. (6) Svensson, S.; Ostberg, T.; Jacobsson, M.; Norstrom, C.; Stefansson, K.; Hallen, D.; Johansson, I. C.; Zachrisson, K.; Ogg, D.; Jendeberg, L. Crystal structure of the heterodimeric complex of LXRα and RXRβ ligand-binding domains in a fully agonistic conformation. EMBO J. 2003, 22, 4625−4633. (7) Nahoum, V.; Perez, E.; Germain, P.; Rodriquez-Barrios, F.; Manzo, F.; Kammerer, S.; Lemaire, G.; Hirsch, O.; Royer, C. A.; Gronemeyer, H.; de Lera, A. R.; Bourguet, W. Modulators of the structural dynamics of the retinoid X receptor to reveal receptor function. Proc. Natl. Acad. Sci. U. S. A. 2007, 104, 17323−17328. (8) Forman, B. M.; Umesono, K.; Chen, J.; Evans, R. M. Unique response pathways are established by allosteric interactions among nuclear hormone receptors. Cell 1995, 81, 541−550. (9) Lala, D. S.; Mukherjee, R.; Schulman, I. G.; Canan Koch, S. S.; Dardashti, L. J.; Nadzan, A. M.; Croston, G. E.; Evans, R. M.; Heyman, R. A. Activation of specific RXR heterodimers by an antagonist of RXR homodimers. Nature 1996, 383, 450−453. 8452

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

Nugent, W. C.; Hermann, T.; Negro-Vilar, A.; Rigas, J. R.; Dmitrovsky, E. A Proof-of-Principle Clinical Trial of Bexarotene in Patients with Non-Small Cell Lung Cancer. Clin. Cancer Res. 2007, 13, 1794−1800. (22) Mukherjee, R.; Davies, P. J. A.; Crombie, D. L.; Bischoff, E. D.; Cesario, R. M.; Jow, L.; Hamanns, L. G.; Boehm, M. F.; Mondon, C. E.; Nadzan, A. M.; Paterniti, J. R.; Heyman, R. A. Sensitization of diabetic and obese mice to insulin by retinoid X receptor agonists. Nature 1997, 386, 407−410. (23) Sherman, S. I.; Gopal, J.; Haugen, B. R.; Chiu, A. C.; Whaley, K.; Nowlakha, P.; Duvic, M. Central hypothyroidism associated with retinoid X receptor-selective ligands. N. Engl. J. Med. 1999, 340, 1075− 1079. (24) Li, D.; Li, T.; Wang, F.; Tian, H.; Samuels, H. H. Functional Evidence for Retinoid X Receptor (RXR) as a Nonsilent Partner in the Thyroid Hormone Receptor/RXR Heterodimer. Mol. Cell. Biol. 2002, 22, 5782−5792. (25) (a) Murthy, S.; Born, E.; Mathur, S. N.; Field, F. J. LXR/RXR activation enhances basolateral efflux of cholesterol in CaCo-2 cells. J. Lipid Res. 2002, 43, 1054−1064. (b) Field, F. J.; Born, E.; Mathur, S. N. LXR/RXR ligand activation enhances basolateral efflux of betasitosterol in CaCo-2 cells. J. Lipid Res. 2004, 45, 905−913. (26) Thacher, S. M.; Standeven, A. M.; Athanikar, J.; Kopper, S.; Castilleja, O.; Escobar, M.; Beard, R. L.; Chandraratna, R. A. S. Receptor Specificity of Retinoid-Induced Epidermal Hyperplasia: Effect of RXR-Selective Agonists and Correlation with Topical Irritation. JPET 1997, 282, 528−534. (27) (a) Cramer, P. E.; Cirrito, J. R.; Wesson, D. W.; Lee, C. Y. D.; Karlo, J. C.; Zinn, A. E.; Casali, B. T.; Restivo, J. L.; Goebel, D. A.; James, M. J.; Brunden, K. R.; Wilson, D. A.; Landreth, G. E. ApoEDirected Therapeutics Rapidly Clear β-Amyloid and Reverse Deficits in AD Mouse Models. Science 2012, 335, 1503−1506. (b) Landreth, G. E.; Cramer, P. E.; Lakner, M. M.; Cirrito, J. R.; Wesson, D. W.; Brunden, K. R.; Wilson, D. A. Response to Comments on ‘ApoEDirected Therapeutics Rapidly Clear β-Amyloid and Reverse Deficits in AD Mouse Models.’. Science 2013, 340, 924. (28) Vuligonda, V.; Thacher, S. M.; Chandraratna, R. A. S. Enantioselective Synthesis of Potent Retinoid X Receptor Ligands: Differential Biological Activities of Individual Antipodes. J. Med. Chem. 2001, 44, 2298−2303. (29) Farmer, L. J.; Marron, K. S.; Canan Koch, S. S.; Hwang, C. K.; Kallel, E. A.; Zhi, L.; Nadzan, A. M.; Robertson, D. W.; Bennani, Y. L. Aza-retinoids as novel retinoid X receptor-specific agonists. Bioorg. Med. Chem. Lett. 2006, 16, 2352−2356. (30) Kagechika, H.; Kawachi, E.; Hashimoto, Y.; Himi, T.; Shudo, K. Retinobenzoic Acids. 1. Structure−Activity Relationships of Aromatic Amides with Retinoidal Activity. J. Med. Chem. 1988, 31, 2182−2192. (31) Michellys, P. Y.; Ardecky, R. J.; Chen, J. H.; Crombie, D. L.; Etgen, G. J.; Faul, M. M.; Faulkner, A. L.; Grese, T. A.; Heyman, R. A.; Karanewsky, D. S.; Klausing, K.; Leibowitz, M. D.; Liu, S.; Mais, D. A.; Mapes, C. M.; Marschke, K. B.; Reifel-Miller, A.; Ogilvie, K. M.; Rungta, D.; Thompson, A. W.; Tyhonas, J. S.; Boehm, M. F. Novel (2E,4E,6Z)-7-(2-Alkoxy-3,5-dialkylbenzene)-3-methylocta-2,4,6-trienoic Acid Retinoid X Receptor Modulators are Active in Models of Type 2 Diabetes. J. Med. Chem. 2003, 46, 2683−2696. (32) Michellys, P.-Y.; Ardecky, R. J.; Chen, J.-H.; D’Arrigo, J.; Grese, T. A.; Karanewsky, D. S.; Leibowitz, M. D.; Liu, S.; Mais, D. A.; Mapes, C. M.; Montrose-Rafizadeh, C.; Ogilvie, K. M.; Reifel-Miller, A.; Rungta, D.; Thompson, A. W.; Tyhonas, J. S.; Boehm, M. F. Design, Synthesis, and Structure−Activity Relationship Studies of Novel 6,7Locked-[7-(2-alkoxy-3,5-dialkylbenzene)-3-methylocta]-2,4,6-trienoic Acids. J. Med. Chem. 2003, 46, 4087−4103. (33) Michellys, P.-Y.; Darrigo, J.; Grese, T. A.; Karanewsky, D. S.; Leibowitz, M. D.; Mais, D. A.; Mapes, C. M.; Reifel-Miller, A.; Rungta, D.; Boehm, M. F. Design, synthesis and structure−activity relationship of novel RXR-selective modulators. Bioorg. Med. Chem. Lett. 2004, 14, 1593−1598. (34) Wagner, C. E.; Jurutka, P. W.; Marshall, P. A.; Groy, T. L.; van der Vaart, A.; Ziller, J. W.; Furmick, J. K.; Graeber, M. E.; Matro, E.; Miguel, B. V.; Tran, I. T.; Kwon, J.; Tedeschi, J. N.; Moosavi, S.;

Danishyar, A.; Philp, J. S.; Khamees, R. O.; Jackson, J. N.; Grupe, D. K.; Badshah, S. L.; Hart, J. W. Modeling, Synthesis and Biological Evaluation of Potential Retinoid X Receptor (RXR) Selective Agonists: Novel Analogues of 4-[1-(3,5,5,8,8-Pentamethyl-5,6,7,8tetrahydro-2-naphthyl)ethynyl]benzoic Acid (Bexarotene). J. Med. Chem. 2009, 52, 5950−5966. (35) Furmick, J. K.; Kaneko, I.; Walsh, A. N.; Yang, J.; Bhogal, J. S.; Gray, G. M.; Baso, J. C.; Browder, D. O.; Prentic, J. L. S.; Montano, L. A.; Huynh, C. C.; Marcus, L. M.; Tsosie, D. G.; Kwon, J. S.; Quezada, A.; Reyes, N. M.; Lemming, B.; Saini, P.; van der Vaart, A.; Groy, T. L.; Marshall, P. A.; Jurutka, P. W.; Wagner, C. E. Modeling, Synthesis and Biological Evaluation of Potential Retinoid X Receptor-Selective Agonists: Novel Halogenated Analogues of 4-[1-(3,5,5,8,8-Pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethynyl]benzoic Acid (Bexarotene). ChemMedChem 2012, 7, 1551−1566. (36) Boehm, M. F.; Zhang, L.; Zhi, L.; McClurg, M. R.; Berger, E.; Wagoner, M.; Mais, D. E.; Suto, C. M.; Davies, P. J. A.; Heyman, R. A.; Nadzan, A. M. Design and Synthesis of Potent Retinoid X Receptor Selective Ligands That Induce Apoptosis in Leukemia Cells. J. Med. Chem. 1995, 38, 3146−3155. (37) Zhang, L.; Badea, B. A.; Enyeart, D.; Berger, E. M.; Mais, D. E.; Boehm, M. F. Synthesis of Isotopically Labeled 4-[1-(3,5,5,8,8Pentamethyl-5,6,7,8-tetrahydro-2-naphthyl)ethenyl]benzoic Acid (LGD1069). J. Labelled Compd. Radiopharm. 1995, 36, 701−712. (38) Faul, M. M.; Ratz, A. M.; Sullivan, K. A.; Trankle, W. G.; Winneroski, L. L. Synthesis of Novel Retinoid X Receptor-Selective Retinoids. J. Org. Chem. 2001, 66, 5772−5782. (39) Santin, E. P.; Germain, P.; Quillard, F.; Khanwalkar, H.; Rodriguez-Barrios, F.; Gronemeyer, H.; de Lera, A. R.; Bourguet, W. Modulating Retinoid X Receptor with a Series of (E)-3-[4-Hydroxy-3(3-alkoxy-5,5,8,8-tetramethyl-5,6,7,8-tetrahydronaphthalen-2-yl)phenyl]acrylic Acids and Their 4-Alkoxy Isomers. J. Med. Chem. 2009, 52, 3150−3158. (40) Gianni, M.; Ponzanelli, I.; Mologni, L.; Reichert, U.; Rambaldi, A.; Terao, M.; Garattini, E. Retinoid-dependent growth inhibition, differentiation and apoptosis in acute promyelocytic leukemia cells. Expression and activation of caspases. Cell Death Differ. 2000, 7, 447− 460. (41) (a) Shapiro, R. H.; Heath, M. J.; Tosylhydrazones, V. Reaction of Tosylhydrazones with Alkyllithium Reagents. A New Olefin Synthesis. J. Am. Chem. Soc. 1967, 89, 5734−5735. (b) Shapiro, R. H.; Lipton, M. F.; Kolonko, K. J.; Buswell, R. L.; Capuano, L. A. Tosylhydrazones and alkyllithium reagents: more on the regiospecificity of the reaction and the trapping of three intermediates. Tetrahedron Lett. 1975, 16, 1811−1814. (42) Miyaura, N.; Yamada, K.; Suzuki, A. A new stereospecific crosscoupling by the palladium-catalyzed reaction of 1-alkenylboranes with 1-alkenyl or 1-alkynyl halides. Tetrahedron Lett. 1979, 20, 3437−3440. (43) Adams, N. D.; Adams, J. L.; Burgess, J. L.; Chaudhari, A. M.; Copeland, R. A.; Donatelli, C. A.; Drewry, D. H.; Fisher, K. E.; Hamajima, T.; Hardwicke, M. A.; Huffman, W. F.; Koretke-Brown, K. K.; Lai, Z. V.; McDonald, O. B.; Nakamura, H.; Newlander, K. A.; Oleykowski, C. A.; Parrish, C. A.; Patrick, D. R.; Plant, R.; Sarpong, M. A.; Sasaki, K.; Schmidt, S. J.; Silva, D. J.; Sutton, D.; Tang, J.; Thompson, C. S.; Tummino, P. J.; Wang, J. C.; Xiang, H.; Yang, J.; Dhanak, D. Discovery of GSK1070916, a Potent and Selective Inhibitor of Aurora B/C Kinase. J. Med. Chem. 2010, 53, 3973−4001. (44) (a) Horner, L.; Hoffmann, H. M. R.; Wippel, H. G. Phosphororganische Verbindungen, XII. Phosphinoxyde als Olefinierungsreagenzien. Chem. Ber. 1958, 91, 61−63. (b) Wadsworth, W. S., Jr.; Emmons, W. D. The Utility of Phosphonate Carbanions in Olefin Synthesis. J. Am. Chem. Soc. 1961, 83, 1733. (45) Lalloyer, F.; Pedersen, T. A.; Gross, B.; Lestavel, S.; Yous, S.; Vallez, E.; Gustafsson, J.-A.; Mandrup, S.; Fievet, C.; Staels, B.; Tailleux, A. Rexinoid Bexarotene Modulates Triglyceride but not Cholesterol Metabolism via Gene-Specific Permissivity of the RXR/ LXR Heterodimer in the Liver. Arterioscler. Thromb. Vasc. Biol. 2009, 29, 1488−1495. 8453

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454

Journal of Medicinal Chemistry

Article

(46) Wan, Y.-J. Y.; An, D.; Cai, Y.; Repa, J. J.; Chen, T. H.-P.; Flores, M.; Postic, C.; Magnuson, M. A.; Chen, J.; Chien, K. R.; French, S.; Mangelsdorf, D. J.; Sucov, H. M. Hepatocyte-Specific Mutation Establishes Retinoid X Receptor α as a Heterodimeric Integrator of Multiple Physiological Processes in the Liver. Mol. Cell. Biol. 2000, 20, 4436−4444. (47) van der Leede, B.-j. M.; van den Brink, C. E.; van der Saag, P. T. Retinoic Acid Receptor and Retinoid X Receptor Expression in Retinoic Acid-Resistant Human Tumor Cell Lines. Mol. Carcinog. 1993, 8, 112−122. (48) Zhang, C.; Hazarika, P.; Ni, X.; Weidner, D. A.; Duvic, M. Induction of Apoptosis by Bexarotene in Cutaneous T-Cell Lymphoma Cells. Clin. Cancer Res. 2002, 8, 1234−1240. (49) Budihardjo, I.; Holt, O.; Lutter, M.; Luo, X.; Wang, X. Biochemical Pathways of Caspase Activation During Apoptosis. Annu. Rev. Cell Dev. Biol. 1999, 15, 269−290. (50) (a) Kotani, H.; Tanabe, H.; Mizukami, H.; Amagaya, S.; Inoue, M. A Naturally Occurring Rexinoid, Honokiol, Can Serve as a Regulator of Various Retinoid X Receptor Heterodimers. Biol. Pharm. Bull. 2012, 35, 1−9. (b) Zou, A.; Elgort, M. G.; Allegretto, E. A. Retinoid X receptor (RXR) ligands activate the human 25hydroxyvitamin D3−24-hydroxylase promoter via RXR heterodimer binding to two vitamin D-responsive elements and elicit additive effects with 1,25-dihydroxyvitamin D3. J. Biol. Chem. 1997, 272, 19027−19034. (51) Hallenbeck, P. L.; Phyillaier, M.; Nikodem, V. M. Divergent effects of 9-cis-retinoic acid receptor on positive and negative thyroid hormone receptor-dependent gene expression. J. Biol. Chem. 1993, 268, 3825−3828. (52) (a) Hong-Qi, Y.; Zhi-Kun, S.; Sheng-Di, C. Current advances in the treatment of Alzheimer’s disease: focus on considerations targeting Aβ and tau. Transl. Neurodegener. 2012, 1, 21. (b) Gross, M. Understanding amyloid and Alzheimer’s disease. Curr. Biol. 2012, R381−R384. (c) Appleby, B. S.; Nacopoulos, D.; Milano, N.; Zhong, K.; Cummings, J. L. A Review: Treatment of Alzheimer’s Disease Discovered in Repurposed Agents. Dementia Geriatr. Cognit. Disord. 2013, 35, 1−22. (53) Mangelsdorf, D. J.; Umesono, K.; Kliewer, S. A.; Borgmeyer, U.; Ong, E. S.; Evans, R. M. A direct repeat in the cellular retinol-binding protein type II gene confers differential regulation by RXR and RAR. Cell 1991, 66, 555−561. (54) Forman, B. M.; Umesono, K.; Chen, J.; Evans, R. M. Unique response pathways are established by allosteric interactions among nuclear hormone receptors. Cell 1995, 81, 541−550. (55) (a) Zimmerman, F. K. Procedures used in the induction of mitotic recombination and mutation in the yeast Saccharomyces cerevisiae. Mutat. Res. 1975, 31, 71−86. (b) Zimmermann, F. K.; Kern, R.; Rasenberger, H. A yeast strain for simultaneous detection of mitotic crossing over, mitotic gene conversion, and reverse mutation. Mutat. Res. 1975, 28, 381−388. (56) Marshall, P. A. Using Saccharomyces cerevisiae to Test the Mutagenicity of Household Compounds: An Open Ended Hypothesis-Driven Teaching Lab. CBE−Life Sci. Educ. 2007, 6, 307−315. (57) Egea, P. F.; Mitschler, A.; Moras, D. Molecular Recognition of Agonist Ligands by RXRs. Mol. Endocrinol. 2002, 16, 987−997. (58) Love, J. D.; Gooch, J. T.; Benko, S.; Li, C.; Nagy, L.; Chatterjee, V. K. K.; Evans, R. M.; Schwabe, J. W. R. The Structural Basis for the Specificity of Retinoid-X Receptor-selective Agonists: New Insights Into the Role of Helix H12. J. Biol. Chem. 2002, 277, 11385−11391. (59) Morris, G. M.; Huey, R.; Lindstrom, W.; Sanner, M. F.; Belew, R. K.; Goodsell, D. S.; Olson, A. J. AutoDock4 and AutoDock Tools4: automated docking with selective receptor flexibility. J. Comput. Chem. 2009, 30, 2785−2791. (60) O’Boyle, N. M.; Banck, M.; James, C. A.; Morley, C.; Vandermeersch, T.; Hutchison, G. R. Open Babel: an open chemical toolbox. J. Cheminf. 2011, 3, 33.

8454

dx.doi.org/10.1021/jm4008517 | J. Med. Chem. 2013, 56, 8432−8454